1
|
Lu HH, Froidevaux C, Biermann I, Kaňková H, Büchner M, Schubert DW, Salehi S, Boccaccini AR. Printable ADA-GEL-based composite inks containing Zn-doped bioactive inorganic fillers for skeletal muscle biofabrication. BIOMATERIALS ADVANCES 2025; 172:214233. [PMID: 40048902 DOI: 10.1016/j.bioadv.2025.214233] [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: 11/10/2024] [Revised: 01/27/2025] [Accepted: 02/13/2025] [Indexed: 03/17/2025]
Abstract
Bioprinting shows significant promise in advancing medical treatments by offering patient-specific solutions for repairing skeletal muscle tissues. Zinc (Zn) is one of the key ions in the human body involved in the development of myogenic cells. This study investigates the integration of Zn-doped bioactive inorganic fillers (BIFs) into alginate-dialdehyde-gelatin (ADA-GEL) as composite ink for bioprinting applications. BIFs were mesoporous calcium-silicate-based bioactive glass nanoparticles with nominal composition: 80% SiO2-X% CaO-Y% ZnO (X = 20, 18, 15, or 10; Y = 0, 2, 5, or 10; mol%). Ion release profiles confirmed that the addition of Zn ions prevented the burst release of Si and Ca ions. The incorporation of BIFs, particularly at higher dopant concentrations, significantly affected the hydrogel swelling and mechanical properties. With increasing concentration of Zn ions (to 5 mol%), the hydrogels exhibited greater dimensional swelling after 24 h of incubation in aqueous solutions, while all compositions lost weight over time after the initial swelling phase. Indirect cell studies demonstrated that 0.1 wt% BIFs extracts, obtained after 24 h-incubation in a cell culture medium, were cytocompatible with C2C12 cells. Furthermore, bioprinted C2C12 cells encapsulated in the bioinks showed a clear increase in cell number after seven days of culture. In particular, cells in the composite inks containing Zn-BIFs exhibited higher spreading, elongation, and alignment than those in pure ADA-GEL, indicating the biological activity provided by the Zn-BIF particles. This study introduces therefore an effective formulation of ADA-GEL-based composite bioinks enriched with Zn-containing nanoparticles for skeletal muscle tissue engineering applications.
Collapse
Affiliation(s)
- Hsuan-Heng Lu
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Clara Froidevaux
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Isabell Biermann
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, 97070 Würzburg, Germany
| | - Hana Kaňková
- FunGlass - Centre for Functional and Surface Functionalized Glass, Alexander Dubček University of Trenčín, 911 50 Trenčín, Slovakia
| | - Margitta Büchner
- Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Dirk W Schubert
- Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Sahar Salehi
- Department of Biomaterials, Faculty of Engineering Science, University of Bayreuth, 95447 Bayreuth, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany.
| |
Collapse
|
2
|
Lee SH, Yoo S, Kim SH, Kim YM, Han SI, Lee H. Nature-inspired surface modification strategies for implantable devices. Mater Today Bio 2025; 31:101615. [PMID: 40115053 PMCID: PMC11925587 DOI: 10.1016/j.mtbio.2025.101615] [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: 12/25/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Medical and implantable devices are essential instruments in contemporary healthcare, improving patient quality of life and meeting diverse clinical requirements. However, ongoing problems such as bacterial colonization, biofilm development, foreign body responses, and insufficient device-tissue adhesion hinder the long-term effectiveness and stability of these devices. Traditional methods to alleviate these issues frequently prove inadequate, necessitating the investigation of nature-inspired alternatives. Biomimetic surfaces, inspired by the chemical and physical principles found in biological systems, present potential opportunities to address these challenges. Recent breakthroughs in manufacturing techniques, including lithography, vapor deposition, self-assembly, and three-dimensional printing, now permit precise control of surface properties at the micro- and nanoscale. Biomimetic coatings can diminish inflammation, prevent bacterial adherence, and enhance stable tissue integration by replicating the antifouling, antibacterial, and adhesive properties observed in creatures such as geckos, mussels, and biological membranes. This review emphasizes the cutting-edge advancements in biomimetic surfaces for medical and implantable devices, outlining their design methodologies, functional results, and prospective clinical applications. Biomimetic coatings, by integrating biological inspiration with advanced surface engineering, have the potential to revolutionize implantable medical devices, providing safer, more lasting, and more effective interfaces for prolonged patient benefit.
Collapse
Affiliation(s)
- Soo-Hwan Lee
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sungjae Yoo
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sung Hoon Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Young-Min Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sang Ihn Han
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyojin Lee
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
- SKKU-KIST, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| |
Collapse
|
3
|
Heltmann‐Meyer S, Detsch R, Hazur J, Kling L, Pechmann S, Kolan RR, Osterloh J, Boccaccini AR, Christiansen S, Geppert CI, Arkudas A, Horch RE, Steiner D. Biofunctionalization of ADA-GEL Hydrogels Based on the Degree of Cross-Linking and Polymer Concentration Improves Angiogenesis. Adv Healthc Mater 2025; 14:e2500730. [PMID: 40095294 PMCID: PMC12023838 DOI: 10.1002/adhm.202500730] [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: 02/16/2025] [Revised: 02/26/2025] [Indexed: 03/19/2025]
Abstract
The creation of bioartificial tissues is a promising option for the reconstruction of large-volume defects. The vascularization of tissue engineering constructs, as well as the material properties of the carrier matrix, are important factors for successful clinical application. In this regard, hydrogels are promising biomaterials, providing an extracellular matrix-like milieu that enables the possibility of cell transplantation and de novo tissue formation. Furthermore, biofunctionalization allows for a certain fine-tuning of angiogenic properties. This study aims to investigate vascularization and tissue formation of highly cross-linked alginate dialdehyde (ADA) and gelatin (GEL). This highly cross-linked network is created using a dural cross-linking mechanism combining ionic (Ca2+ ions) and enzymatic (human transglutaminase (hTG)) cross-linking, resulting in reduced swelling and moderate degradation rates. Vascularization of the ADA-GEL-hTG constructs is induced surgically using arteriovenous (AV) loops. Biocompatibility, tissue formation, and vascularization are analyzed by histology and X-ray microscopy. After only 2 weeks, vascularization of the ADA-GEL-hTG constructs is already present. After 4 weeks, both de novo tissue formation and vascularization of the ADA-GEL-hTG matrix increase. In conclusion, ADA-GEL-hTG-based hydrogels are shown to be promising scaffold materials for tissue engineering applications.
Collapse
Affiliation(s)
- Stefanie Heltmann‐Meyer
- Department of Plastic and Hand SurgeryUniversity Hospital of ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| | - Rainer Detsch
- Institute of BiomaterialsUniversity of Erlangen‐Nürnberg91058ErlangenGermany
| | - Jonas Hazur
- Institute of BiomaterialsUniversity of Erlangen‐Nürnberg91058ErlangenGermany
| | - Lasse Kling
- Institute for Nanotechnology and Correlative Microscopy gGmbH (INAM gGmbH)91301ForchheimGermany
| | - Sabrina Pechmann
- Department for Correlative Microscopy and Materials DataFraunhofer Institute for Ceramic Technologies and Systems (IKTS)91301ForchheimGermany
| | - Rajkumar Reddy Kolan
- Institute for Nanotechnology and Correlative Microscopy gGmbH (INAM gGmbH)91301ForchheimGermany
| | - Justus Osterloh
- Department of Plastic and Hand SurgeryUniversity Hospital of ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)91054ErlangenGermany
- Department of Plastic and Hand SurgeryUniversity of Freiburg Medical Center79106FreiburgGermany
| | - Aldo R. Boccaccini
- Institute of BiomaterialsUniversity of Erlangen‐Nürnberg91058ErlangenGermany
| | - Silke Christiansen
- Department for Correlative Microscopy and Materials DataFraunhofer Institute for Ceramic Technologies and Systems (IKTS)91301ForchheimGermany
- Fachbereich PhysikFreie Universität Berlin (FU Berlin)14195BerlinGermany
| | - Carol I. Geppert
- Institute of PathologyUniversity Hospital of ErlangenFriedrich‐Alexander‐UniversitätErlangen‐Nürnberg (FAU)91054ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)University Hospital ErlangenFAU Erlangen‐Nuremberg91054ErlangenGermany
| | - Andreas Arkudas
- Department of Plastic and Hand SurgeryUniversity Hospital of ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| | - Raymund E. Horch
- Department of Plastic and Hand SurgeryUniversity Hospital of ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)91054ErlangenGermany
| | - Dominik Steiner
- Department of Plastic and Hand SurgeryUniversity Hospital of ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)91054ErlangenGermany
- Department of HandPlasticReconstructiveand Burn SurgeryBG Trauma ClinicUniversity of Tübingen72076TübingenGermany
| |
Collapse
|
4
|
Faber J, Hinrichsen J, Soufivand AA, Lu HH, Rosenberger T, Karakaya E, Detsch R, Boccaccini AR, Budday S. Tuning the mechanical properties of alginate dialdehyde-gelatin (ADA-GEL) bioinks for bioprinting approaches by varying the degree of oxidation. J Mech Behav Biomed Mater 2025; 163:106871. [PMID: 39764923 DOI: 10.1016/j.jmbbm.2024.106871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 02/08/2025]
Abstract
Extrusion-based 3D bioprinting is one of the most promising and widely used technologies in bioprinting. However, the development of bioprintable, biocompatible bioinks with tailored mechanical and biological properties remains a major challenge in this field. Alginate dialdehyde-gelatin (ADA-GEL) hydrogels face these difficulties and enable to tune the mechanical properties depending on the degree of oxidation (% DO) of ADA. Here, we present a holistic approach for characterizing the influence of the % DO on the mechanical properties of ADA-GEL hydrogels under multiple loading modes, compression, tension, and torsional shear in the large-strain regime. We evaluate complex mechanical characteristics including nonlinearity, hysteresis, conditioning, and stress relaxation. We calibrate hyperelastic material models to determine the corresponding material parameters inversely. Our results confirm that decreasing the % DO of ionically crosslinked ADA-GEL hydrogels leads to an increase in stiffness, more distinct nonlinearity, more pronounced hysteresis, and minor preconditioning effects, while the relaxation behavior is slightly affected. The fabrication technique - molding or printing - does only slightly affect the complex mechanical properties and stress relaxation behavior. Ionically and enzymatically dual-crosslinked ADA-GEL hydrogels showed higher stresses during cyclic loading and less viscous effects during stress relaxation in all three loading modes. We conclude that the % DO and the crosslinking procedure are crucial parameters to tune the mechanical behavior of ADA-GEL hydrogels. Careful choice of these parameters might facilitate the fabrication of biomaterials that closely mimic the properties of native tissues for advanced tissue engineering applications.
Collapse
Affiliation(s)
- Jessica Faber
- Institute of Continuum Mechanics and Biomechanics, Department of Mechanical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 90762 Fürth, Germany
| | - Jan Hinrichsen
- Institute of Continuum Mechanics and Biomechanics, Department of Mechanical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 90762 Fürth, Germany
| | - Anahita Ahmadi Soufivand
- Institute of Continuum Mechanics and Biomechanics, Department of Mechanical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 90762 Fürth, Germany
| | - Hsuan-Heng Lu
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91056 Erlangen, Germany
| | - Tanja Rosenberger
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91056 Erlangen, Germany
| | - Emine Karakaya
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91056 Erlangen, Germany
| | - Rainer Detsch
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91056 Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91056 Erlangen, Germany
| | - Silvia Budday
- Institute of Continuum Mechanics and Biomechanics, Department of Mechanical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 90762 Fürth, Germany.
| |
Collapse
|
5
|
Bagewadi S, Rajendran M, Ganapathisankarakrishnan A, Budharaju H, Sethuraman S, Sundaramurthi D. Preparation of thermoresponsive & enzymatically crosslinkable gelatin-gellan gum bioink: A protein-polysaccharide hydrogel for 3D bioprinting of complex soft tissues. Int J Biol Macromol 2025; 295:139563. [PMID: 39788240 DOI: 10.1016/j.ijbiomac.2025.139563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/29/2024] [Accepted: 01/05/2025] [Indexed: 01/12/2025]
Abstract
Developing superior bioinks present several challenges in achieving ideal properties such as biocompatibility, viscosity, degradation rates & mechanical properties which are required to make functional tissue constructs. Various attempts have been made to prepare excellent bioink compositions that are suitable to address the above challenges. Herein, a versatile combination of gelatin (GL) - gellan gum (GG) bioink was successfully formulated & the bioink 7.5GL/2GG was found to be ideal for printing complex and highly intricate structures with excellent shape fidelity. Two different crosslinkers namely transglutaminase (TG) and calcium chloride (CaCl2) were utilized for crosslinking. The rheological properties of GL/GG bioink indicated that TG and dual (TG + CaCl2) crosslinked constructs had storage modulus equivalent to the that of native skin. Direct and indirect cytotoxicity assays revealed that the developed constructs were cytocompatible as well as hemocompatible. The 3D bioprinted GL/GG constructs crosslinked with only TG showed better cell viability, proliferation, cell spreading and wound healing efficiency in vitro compared to dual crosslinked constructs. In conclusion, TG crosslinking of 7.5GL/2GG bioink was ideal for bioprinting of skin tissue constructs for regenerative medicine applications. By altering the concentrations & printing conditions, this bioink may be tuned for other soft tissue engineering applications.
Collapse
Affiliation(s)
- Shambhavi Bagewadi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Madhumathi Rajendran
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Aiswarya Ganapathisankarakrishnan
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India.
| |
Collapse
|
6
|
Bider F, Gunnella C, Reh JT, Clejanu CE, Kuth S, Beltrán AM, Boccaccini AR. Enhancing alginate dialdehyde-gelatin (ADA-GEL) based hydrogels for biofabrication by addition of phytotherapeutics and mesoporous bioactive glass nanoparticles (MBGNs). J Biomater Appl 2025; 39:524-556. [PMID: 39305217 PMCID: PMC11707976 DOI: 10.1177/08853282241280768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
This study explores the 3D printing of alginate dialdehyde-gelatin (ADA-GEL) inks incorporating phytotherapeutic agents, such as ferulic acid (FA), and silicate mesoporous bioactive glass nanoparticles (MBGNs) at two different concentrations. 3D scaffolds with bioactive properties suitable for bone tissue engineering (TE) were obtained. The degradation and swelling behaviour of films and 3D printed scaffolds indicated an accelerated trend with increasing MBGN content, while FA appeared to stabilize the samples. Determination of the degree of crosslinking validated the increased stability of hydrogels due to the addition of FA and 0.1% (w/v) MBGNs. The incorporation of MBGNs not only improved the effective moduli and conferred bioactive properties through the formation of hydroxyapatite (HAp) on the surface of ADA-GEL-based samples but also enhanced VEGF-A expression of MC3T3-E1 cells. The beneficial impact of FA and low concentrations of MBGNs in ADA-GEL-based inks for 3D (bio)printing applications was corroborated through various printing experiments, resulting in higher printing resolution, as also confirmed by rheological measurements. Cytocompatibility investigations revealed enhanced MC3T3-E1 cell activity and viability. Furthermore, the presence of mineral phases, as confirmed by an in vitro biomineralization assay, and increased ALP activity after 21 days, attributed to the addition of FA and MBGNs, were demonstrated. Considering the acquired structural and biological properties, along with efficient drug delivery capability, enhanced biological activity, and improved 3D printability, the newly developed inks exhibit promising potential for biofabrication and bone TE.
Collapse
Affiliation(s)
- Faina Bider
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Chiara Gunnella
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Jana T Reh
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Corina-Elena Clejanu
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Sonja Kuth
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Ana M Beltrán
- Departamento de Ingeniería y Ciencia de los Materiales y del Transporte. Escuela Politécnica Superior, Virgen de África 7, Universidad de Sevilla, Seville (Spain)
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
7
|
Coban B, Baskurt M, Sahin H, Arslan-Yildiz A. Development of Mg-Alginate Based Self Disassociative Bio-Ink for Magnetic Bio-Patterning of 3D Tumor Models. Macromol Biosci 2025; 25:e2400339. [PMID: 39555906 DOI: 10.1002/mabi.202400339] [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: 07/16/2024] [Revised: 09/23/2024] [Indexed: 11/19/2024]
Abstract
Alginate forms a hydrogel via physical cross-linking with divalent cations. In literature, Ca2+ is mostly utilized due to strong interactions but additional procedures are required to disassociate Ca-alginate hydrogels. On the other hand, Mg-alginate hydrogels disassociate spontaneously, which might benefit certain applications. This study introduces Mg-alginate as the main component of a bio-ink for the first time to obtain 3D tumor models by magnetic bio-patterning technique. The bio-ink contains magnetic nanoparticles (MNPs) for magnetic manipulation, Mg-alginate hydrogel as a sacrificial material, and cells. The applicability of the methodology is tested for the formation of 3D tumor models using HeLa, SaOS-2, and SH-SY5Y cells. Long-term cultures are examined by Live/dead and MTT analysis and revealed high cell viability. Subsequently, Collagen and F-actin expressions are observed successfully in 3D tumor models. Finally, the anti-cancer drug Doxorubicin (DOX) effect is investigated on 3D tumor models, and IC50 values is calculated to assess the drug response. As a result, significantly higher drug resistance is observed for bio-patterned 3D tumor models up to tenfold compared to 2D control. Overall, Mg-alginate hydrogel is successfully used to form bio-patterned 3D tumor models, and the applicability of the model is shown effectively, especially as a drug screening platform.
Collapse
Affiliation(s)
- Basak Coban
- Department of Bioengineering, Izmir Institute of Technology, Izmir, 35430, Turkey
| | - Mehmet Baskurt
- Department of Photonics, Izmir Institute of Technology, Izmir, 35430, Turkey
| | - Hasan Sahin
- Department of Photonics, Izmir Institute of Technology, Izmir, 35430, Turkey
| | - Ahu Arslan-Yildiz
- Department of Bioengineering, Izmir Institute of Technology, Izmir, 35430, Turkey
| |
Collapse
|
8
|
Zhang X, Yang C, Zeng X, Li G. A bioactive composite sponge based on biomimetic collagen fibril and oxidized alginate for noncompressible hemorrhage and wound healing. Carbohydr Polym 2024; 343:122409. [PMID: 39174076 DOI: 10.1016/j.carbpol.2024.122409] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 08/24/2024]
Abstract
The study focuses on developing a bioactive shape memory sponge to address the urgent demand for short-term rapid hemostasis and long-term wound healing in noncompressible hemorrhage cases. A composite sponge was created by spontaneously generating pores and double cross-linking under mild conditions using biomimetic collagen fibril (BCF) and oxidized alginate (OA) as natural backbone, combined with an inert calcium source (Ca) from CaCO3-GDL slow gelation mechanism. The optimized BCF/OACa (5/5) sponge efficiently absorbed blood after compression and recovered to its original state within 11.2 ± 1.3 s, achieving physical hemostatic mechanism. The composite sponge accelerated physiological coagulation by promoting platelet adhesion and activation through BCF, as well as enhancing endogenous and exogenous hemostatic pathways by Ca2+. Compared to commercial PVA expanding hemostatic sponge, the composite sponge reduced bleeding volume and shortened hemostasis time in rat liver injury pick and perforation wound models. Additionally, it stimulated fibroblast migration and differentiation, thus promoting wound healing. It is biodegradable with low inflammatory response and promotes granulation tissue regeneration. In conclusion, this biocomposite sponge provides multiple hemostatic pathways and biochemical support for wound healing, is biologically safe and easy to fabricate, process and use, with significant potential for clinical translation and application.
Collapse
Affiliation(s)
- Xiaoxia Zhang
- Key Laboratory of Leather Chemistry and Engineering, Ministry of Education, Sichuan University, Chengdu 610065, PR China; National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| | - Changkai Yang
- Key Laboratory of Leather Chemistry and Engineering, Ministry of Education, Sichuan University, Chengdu 610065, PR China; National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| | - Xingling Zeng
- Key Laboratory of Leather Chemistry and Engineering, Ministry of Education, Sichuan University, Chengdu 610065, PR China; National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| | - Guoying Li
- Key Laboratory of Leather Chemistry and Engineering, Ministry of Education, Sichuan University, Chengdu 610065, PR China; National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China.
| |
Collapse
|
9
|
Abroug N, Schöbel L, Boccaccini AR, Seitz H. Quantitative Macromolecular Modeling Assay of Biopolymer-Based Hydrogels. Gels 2024; 10:676. [PMID: 39590032 PMCID: PMC11594088 DOI: 10.3390/gels10110676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
The rubber elasticity theory has been lengthily applied to several polymeric hydrogel substances and upgraded from idealistic models to consider imperfections in the polymer network. The theory relies solely on hyperelastic material models in order to provide a description of the elastic polymer network. While this is also applicable to polymer gels, such hydrogels are rather characterized by their water content and visco-elastic mechanical properties. In this work, we applied rubber elasticity constitutive models through hyperelastic parameter identification of hydrogels based on their stress-strain response to compression. We further performed swelling experiments and determined the intrinsic properties, i.e., density, of the specimens and their components. Additionally, we estimated their equilibrium swelling and employed it in the swelling-equilibrium theory in order to determine the polymer-solvent interaction parameter of each hydrogel with regard to cross-linking. Our results show that the average mesh size obtained from the rubber elasticity theory can be regarded as a concentration-dependent characteristic length of the hydrogel's network and couples the non-linear elastic response to the specimens' inherent visco-elasticity through hysteresis as a quantifier of energy dissipation under large deformation.
Collapse
Affiliation(s)
- Nada Abroug
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18059 Rostock, Germany;
| | - Lisa Schöbel
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich Alexander-University Erlangen-Nuremberg, 91058 Erlangen, Germany; (L.S.); (A.R.B.)
| | - Aldo R. Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich Alexander-University Erlangen-Nuremberg, 91058 Erlangen, Germany; (L.S.); (A.R.B.)
| | - Hermann Seitz
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18059 Rostock, Germany;
- Department Life, Light & Matter, Interdisciplinary Faculty, University of Rostock, 18059 Rostock, Germany
| |
Collapse
|
10
|
Kashi M, Nazarpak MH, Nourmohammadi J, Moztarzadeh F. Study the effect of different concentrations of polydopamine as a secure and bioactive crosslinker on dual crosslinking of oxidized alginate and gelatin wound dressings. Int J Biol Macromol 2024; 277:134199. [PMID: 39069056 DOI: 10.1016/j.ijbiomac.2024.134199] [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: 12/02/2023] [Revised: 07/09/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Alginate hydrogels are commonly used in wound care due to their ability to maintain a moist environment, absorb fluids, and aid wound healing. However, their stability and mechanical properties can sometimes limit their effectiveness. This study explores a new approach by creating a dual network system of oxidized alginate and gelatin hydrogel crosslinked with polydopamine in a single step, with the goal of improving the mechanical properties of these hydrogels. The unique aspect of this research is the comprehensive examination of different polydopamine concentrations in dual crosslinking systems. First, alginate was modified with sodium periodate to create additional active groups on its backbone, and various polydopamine concentrations were then tested to assess their impact on the dual crosslinking network and hydrogel properties. The study involved a range of tests, including FTIR, H-NMR, SEM, gelation time, rheology, adhesion, antioxidant activity, swelling ratio, weight loss, drug release, and cell viability. The addition of polydopamine was found to enhance the crosslinking density (0.859 × 109 mol.cm-3). Additionally, the results indicated improvements in properties such as reduced weight loss, enhanced antioxidant and adhesive qualities, and better mechanical properties (2240 kPa). However, the optimal concentration of polydopamine must be determined to achieve the best properties for a wound dressing. Excessive polydopamine can increase the space between polymer chains, leading to a reduction in crosslinking density and storage modulus. Nevertheless, it can also increase the swelling ratio, degradation rate, pore size, porosity, antioxidant activity, and dopamine release. Therefore, identifying the optimal concentration for a functional hydrogel is crucial. Notably, the hydrogel containing 0.5 mg.mL-1 polydopamine exhibited outstanding cell viability (108 % on the third day), swelling capacity (480 %), storage modulus (2240 kPa), gelation time (3 min), antioxidant activity (42.27 %), and skin adherence (11 kPa), making it an optimal choice for advanced wound management. According to the findings, it is emphasized that the application of this particular hydrogel expedites wound healing, as indicated by wound closure and histological studies. ABBREVIATIONS.
Collapse
Affiliation(s)
- Mana Kashi
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran Polytechnic, Iran
| | - Masoumeh Haghbin Nazarpak
- New Technologies Research Center (NTRC), Amirkabir University of Technology, Tehran Pochronichnic, Iran.
| | - Jhamak Nourmohammadi
- School of Life Sciences Engineering, College of Interdisciplinary Sciences and Technology, University of Tehran, Tehran, Iran.
| | - Fathollah Moztarzadeh
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran Polytechnic, Iran
| |
Collapse
|
11
|
Solhtalab E, Nikokar I, Mojtahedi A, Shokri R, Karimian P, Mahdavi E, Faezi S. Encapsulation of Mentha aquatica methanol extract in alginate hydrogel promotes wound healing in a murine model of Pseudomonas aeruginosa burn infection. Int J Biol Macromol 2024; 280:135920. [PMID: 39322165 DOI: 10.1016/j.ijbiomac.2024.135920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 09/01/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Burn injuries are the fourth most prevalent devastating form of trauma worldwide. Among the most extensively explored materials, composite dressings with alginate loaded with herbal extract can be mentioned. This research aimed to develop a sodium alginate (SA)-based hydrogel encapsulated with Mentha aquatica (MA) methanol extract and investigate its therapeutic efficacy in the infected burn mouse model. SEM, FTIR, in vitro extract release, HPLC, mechanical test, contact angle, swelling, degradability, and temperature response properties were used to analyze the hydrogel scaffold's physicochemical structure. Additionally, the antibacterial activity and MIC level of the extract, cell cytotoxicity, and macroscopic and microscopic analysis of the wound healing process were done using Masson's trichrome and hematoxylin-eosin staining. The physicochemical properties of the SA hydrogel encapsulated with MA extract were verified. The lowest inhibitory dose of the extract was determined to be 12.5 mg/ml. Application of SA/MA hydrogel to localized wounds of deep third-degree burns demonstrated faster tissue regeneration, collagen recovery, and eradication of bacterial infection. This research focused on the design and the preparation of a novel and effective biomaterials-based medical product, which has the potential to rehabilitate infected and injured skin tissue; therefore, it can be a promising candidate for wound dressing applications.
Collapse
Affiliation(s)
- Elahe Solhtalab
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Iraj Nikokar
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Mojtahedi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Shokri
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Paridokht Karimian
- Department of Pathology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Edris Mahdavi
- Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sobhan Faezi
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
12
|
Garcia-Garcia A, Muñana-González S, Lanceros-Mendez S, Ruiz-Rubio L, Alvarez LP, Vilas-Vilela JL. Biodegradable Natural Hydrogels for Tissue Engineering, Controlled Release, and Soil Remediation. Polymers (Basel) 2024; 16:2599. [PMID: 39339063 PMCID: PMC11435712 DOI: 10.3390/polym16182599] [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: 07/22/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
This article provides insights into hydrogels of the most promising biodegradable natural polymers and their mechanisms of degradation, highlighting the different possibilities of controlling hydrogel degradation rates. Since biodegradable hydrogels can be designed as scaffolding materials to mimic the physical and biochemical properties of natural tissues, these hydrogels have found widespread application in the field of tissue engineering and controlled release. In the same manner, their potential as water reservoirs, macro- and microelement carriers, or matrixes for the selective adsorption of pollutants make them excellent candidates for sustainable soil amendment solutions. Accordingly, this article summarizes the recent advances in natural biodegradable hydrogels in the fields of tissue engineering, controlled release, and soil remediation, emphasizing the new opportunities that degradability and its tunability offer for the design and applicability of hydrogels.
Collapse
Affiliation(s)
- Ane Garcia-Garcia
- Macromolecular Chemistry Group (LABQUIMAC), Physical Chemistry Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| | - Sara Muñana-González
- Macromolecular Chemistry Group (LABQUIMAC), Physical Chemistry Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Senentxu Lanceros-Mendez
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Leire Ruiz-Rubio
- Macromolecular Chemistry Group (LABQUIMAC), Physical Chemistry Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| | - Leyre Perez Alvarez
- Macromolecular Chemistry Group (LABQUIMAC), Physical Chemistry Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| | - José Luis Vilas-Vilela
- Macromolecular Chemistry Group (LABQUIMAC), Physical Chemistry Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| |
Collapse
|
13
|
Rashidi S, Bagherpour G, Abbasi‐Malati Z, Khosrowshahi ND, Chegeni SA, Roozbahani G, Lotfimehr H, Sokullu E, Rahbarghazi R. Endothelial progenitor cells for fabrication of engineered vascular units and angiogenesis induction. Cell Prolif 2024; 57:e13716. [PMID: 39051852 PMCID: PMC11503262 DOI: 10.1111/cpr.13716] [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: 05/23/2024] [Revised: 06/21/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024] Open
Abstract
The promotion of vascularization and angiogenesis in the grafts is a crucial phenomenon in the healing process and tissue engineering. It has been shown that stem cells, especially endothelial progenitor cells (EPCs), can stimulate blood vessel formation inside the engineered hydrogels after being transplanted into the target sites. The incorporation of EPCs into the hydrogel can last the retention time, long-term survival, on-target delivery effects, migration and differentiation into mature endothelial cells. Despite these advantages, further modifications are mandatory to increase the dynamic growth and angiogenesis potential of EPCs in in vitro and in vivo conditions. Chemical modifications of distinct composites with distinct physical properties can yield better regenerative potential and angiogenesis during several pathologies. Here, we aimed to collect recent findings related to the application of EPCs in engineered vascular grafts and/or hydrogels for improving vascularization in the grafts. Data from the present article can help us in the application of EPCs as valid cell sources in the tissue engineering of several ischemic tissues.
Collapse
Affiliation(s)
- Somayyeh Rashidi
- Department of Medical Biotechnology, Faculty of MedicineZanjan University of Medical SciencesZanjanIran
| | - Ghasem Bagherpour
- Department of Medical Biotechnology, Faculty of MedicineZanjan University of Medical SciencesZanjanIran
- Zanjan Pharmaceutical Biotechnology Research CenterZanjan University of Medical SciencesZanjanIran
| | - Zahra Abbasi‐Malati
- Student Research CenterTabriz University of Medical SciencesTabrizIran
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
| | | | - Sara Aghakhani Chegeni
- Department of Clinical Biochemistry and Laboratory MedicineTabriz University of Medical SciencesTabrizIran
| | - Golbarg Roozbahani
- Department of Plant, Cell and Molecular Biology, Faculty of Natural SciencesUniversity of TabrizTabrizIran
| | - Hamid Lotfimehr
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
| | - Emel Sokullu
- Research Center for Translational Medicine (KUTTAM)Koç UniversityIstanbulTurkey
- Biophysics DepartmentKoç University School of MedicineIstanbulTurkey
| | - Reza Rahbarghazi
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran
- Department of Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
14
|
Zhanbassynova A, Mukasheva F, Abilev M, Berillo D, Trifonov A, Akilbekova D. Impact of Hydroxyapatite on Gelatin/Oxidized Alginate 3D-Printed Cryogel Scaffolds. Gels 2024; 10:406. [PMID: 38920952 PMCID: PMC11203254 DOI: 10.3390/gels10060406] [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: 05/30/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Fabrication of scaffolds via 3D printing is a promising approach for tissue engineering. In this study, we combined 3D printing with cryogenic crosslinking to create biocompatible gelatin/oxidized alginate (Gel/OxAlg) scaffolds with large pore sizes, beneficial for bone tissue regeneration. To enhance the osteogenic effects and mechanical properties of these scaffolds, we evaluated the impact of hydroxyapatite (HAp) on the rheological characteristics of the 2.86% (1:1) Gel/OxAlg ink. We investigated the morphological and mechanical properties of scaffolds with low, 5%, and high 10% HAp content, as well as the resulting bio- and osteogenic effects. Scanning electron microscopy revealed a reduction in pore sizes from 160 to 180 µm (HAp-free) and from 120 to 140 µm for both HAp-containing scaffolds. Increased stability and higher Young's moduli were measured for 5% and 10% HAp (18 and 21 kPa, respectively) compared to 11 kPa for HAp-free constructs. Biological assessments with mesenchymal stem cells indicated excellent cytocompatibility and osteogenic differentiation in all scaffolds, with high degree of mineralization in HAp-containing constructs. Scaffolds with 5% HAp exhibited improved mechanical characteristics and shape fidelity, demonstrated positive osteogenic impact, and enhanced bone tissue formation. Increasing the HAp content to 10% did not show any advantages in osteogenesis, offering a minor increase in mechanical strength at the cost of significantly compromised shape fidelity.
Collapse
Affiliation(s)
- Ainur Zhanbassynova
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan; (A.Z.)
| | - Fariza Mukasheva
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan; (A.Z.)
| | - Madi Abilev
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan; (A.Z.)
| | - Dmitriy Berillo
- Department of Chemistry and Biochemical Engineering, Satbayev University, Almaty 050013, Kazakhstan
| | - Alexander Trifonov
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan; (A.Z.)
| | - Dana Akilbekova
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan; (A.Z.)
| |
Collapse
|
15
|
Ege D, Boccaccini AR. Investigating the Effect of Processing and Material Parameters of Alginate Dialdehyde-Gelatin (ADA-GEL)-Based Hydrogels on Stiffness by XGB Machine Learning Model. Bioengineering (Basel) 2024; 11:415. [PMID: 38790283 PMCID: PMC11117982 DOI: 10.3390/bioengineering11050415] [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: 03/05/2024] [Revised: 03/26/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
To address the limitations of alginate and gelatin as separate hydrogels, partially oxidized alginate, alginate dialdehyde (ADA), is usually combined with gelatin to prepare ADA-GEL hydrogels. These hydrogels offer tunable properties, controllable degradation, and suitable stiffness for 3D bioprinting and tissue engineering applications. Several processing variables affect the final properties of the hydrogel, including degree of oxidation, gelatin content and type of crosslinking agent. In addition, in 3D-printed structures, pore size and the possible addition of a filler to make a hydrogel composite also affect the final physical and biological properties. This study utilized datasets from 13 research papers, encompassing 33 unique combinations of ADA concentration, gelatin concentration, CaCl2 and microbial transglutaminase (mTG) concentrations (as crosslinkers), pore size, bioactive glass (BG) filler content, and one identified target property of the hydrogels, stiffness, utilizing the Extreme Boost (XGB) machine learning algorithm to create a predictive model for understanding the combined influence of these parameters on hydrogel stiffness. The stiffness of ADA-GEL hydrogels is notably affected by the ADA to GEL ratio, and higher gelatin content for different ADA gel concentrations weakens the scaffold, likely due to the presence of unbound gelatin. Pore size and the inclusion of a BG particulate filler also have a significant impact on stiffness; smaller pore sizes and higher BG content lead to increased stiffness. The optimization of ADA-GEL composition and the inclusion of BG fillers are key determinants to tailor the stiffness of these 3D printed hydrogels, as found by the analysis of the available data.
Collapse
Affiliation(s)
- Duygu Ege
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany;
- Institute of Biomedical Engineering, Bogazici University, Rasathane St., Kandilli, 34684 İstanbul, Turkey
| | - Aldo R. Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany;
| |
Collapse
|
16
|
Charoenchokpanich W, Muangrod P, Roytrakul S, Rungsardthong V, Wonganu B, Charoenlappanit S, Casanova F, Thumthanaruk B. Exploring the Model of Cefazolin Released from Jellyfish Gelatin-Based Hydrogels as Affected by Glutaraldehyde. Gels 2024; 10:271. [PMID: 38667690 PMCID: PMC11048929 DOI: 10.3390/gels10040271] [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: 03/23/2024] [Revised: 04/11/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
Due to its excellent biocompatibility and ease of biodegradation, jellyfish gelatin has gained attention as a hydrogel. However, hydrogel produced from jellyfish gelatin has not yet been sufficiently characterized. Therefore, this research aims to produce a jellyfish gelatin-based hydrogel. The gelatin produced from desalted jellyfish by-products varied with the part of the specimen and extraction time. Hydrogels with gelatin: glutaraldehyde ratios of 10:0.25, 10:0.50, and 10:1.00 (v/v) were characterized, and their cefazolin release ability was determined. The optimal conditions for gelatin extraction and chosen for the development of jellyfish hydrogels (JGel) included the use of the umbrella part of desalted jellyfish by-products extracted for 24 h (WU24), which yielded the highest gel strength (460.02 g), viscosity (24.45 cP), gelling temperature (12.70 °C), and melting temperature (22.48 °C). The quantities of collagen alpha-1(XXVIII) chain A, collagen alpha-1(XXI) chain, and collagen alpha-2(IX) chain in WU24 may influence its gel properties. Increasing the glutaraldehyde content in JGel increased the gel fraction by decreasing the space between the protein chains and gel swelling, as glutaraldehyde binds with lateral amino acid residues and produces a stronger network. At 8 h, more than 80% of the cefazolin in JGel (10:0.25) was released, which was higher than that released from bovine hydrogel (52.81%) and fish hydrogel (54.04%). This research is the first report focused on the production of JGel using glutaraldehyde as a cross-linking agent.
Collapse
Affiliation(s)
- Wiriya Charoenchokpanich
- Department of Agro-Industrial, Food, and Environmental Technology, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand; (W.C.); (P.M.); (V.R.)
| | - Pratchaya Muangrod
- Department of Agro-Industrial, Food, and Environmental Technology, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand; (W.C.); (P.M.); (V.R.)
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (S.R.); (S.C.)
| | - Vilai Rungsardthong
- Department of Agro-Industrial, Food, and Environmental Technology, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand; (W.C.); (P.M.); (V.R.)
- Food and Agro-Industry Research Center, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand
- Center for Food Industry Innovation Technology, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand
| | - Benjamaporn Wonganu
- Department of Biotechnology, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand;
| | - Sawanya Charoenlappanit
- Functional Proteomics Technology Laboratory, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (S.R.); (S.C.)
| | - Federico Casanova
- Research Group for Food Production Engineering, National Food Institute, Technical University of Denmark, 28000 Kongens Lyngby, Denmark;
| | - Benjawan Thumthanaruk
- Department of Agro-Industrial, Food, and Environmental Technology, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand; (W.C.); (P.M.); (V.R.)
- Food and Agro-Industry Research Center, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand
- Center for Food Industry Innovation Technology, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand
| |
Collapse
|
17
|
Kim M, Schöbel L, Geske M, Boccaccini AR, Ghorbani F. Bovine serum albumin-modified 3D printed alginate dialdehyde-gelatin scaffolds incorporating polydopamine/SiO 2-CaO nanoparticles for bone regeneration. Int J Biol Macromol 2024; 264:130666. [PMID: 38453119 DOI: 10.1016/j.ijbiomac.2024.130666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Three-dimensional (3D) printing allows precise manufacturing of bone scaffolds for patient-specific applications and is one of the most recently developed and implemented technologies. In this study, bilayer and multimaterial alginate dialdehyde-gelatin (ADA-GEL) scaffolds incorporating polydopamine (PDA)/SiO2-CaO nanoparticle complexes were 3D printed using a pneumatic extrusion-based 3D printing technology and further modified on the surface with bovine serum albumin (BSA) for application in bone regeneration. The morphology, chemistry, and in vitro bioactivity of PDA/SiO2-CaO nanoparticle complexes were characterized (n = 3) and compared with those of mesoporous SiO2-CaO nanoparticles. Successful deposition of the PDA layer on the surface of the SiO2-CaO nanoparticles allowed better dispersion in a liquid medium and showed enhanced bioactivity. Rheological studies (n = 3) of ADA-GEL inks consisting of PDA/SiO2-CaO nanoparticle complexes showed results that may indicate better injectability and printability behavior compared to ADA-GEL inks incorporating unmodified nanoparticles. Microscopic observations of 3D printed scaffolds revealed that PDA/SiO2-CaO nanoparticle complexes introduced additional topography onto the surface of 3D printed scaffolds. Additionally, the modified scaffolds were mechanically stable and elastic, closely mimicking the properties of natural bone. Furthermore, protein-coated bilayer scaffolds displayed controllable absorption and biodegradation, enhanced bioactivity, MC3T3-E1 cell adhesion, proliferation, and higher alkaline phosphatase (ALP) activity (n = 3) compared to unmodified scaffolds. Consequently, the present results confirm that ADA-GEL scaffolds incorporating PDA/SiO2-CaO nanoparticle complexes modified with BSA offer a promising approach for bone regeneration applications.
Collapse
Affiliation(s)
- MinJoo Kim
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany; Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Lisa Schöbel
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
| | - Michael Geske
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany; Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Martensstraße 7, 91058 Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany.
| | - Farnaz Ghorbani
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany; Department of Translational Health Sciences, University of Bristol, Bristol BS1 3NY, UK.
| |
Collapse
|
18
|
Willems C, Qi F, Trutschel ML, Groth T. Functionalized Gelatin/Polysaccharide Hydrogels for Encapsulation of Hepatocytes. Gels 2024; 10:231. [PMID: 38667650 PMCID: PMC11048940 DOI: 10.3390/gels10040231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Liver diseases represent a considerable burden to patients and healthcare systems. Hydrogels play an important role in the engineering of soft tissues and may be useful for embedding hepatocytes for different therapeutic interventions or the development of in vitro models to study the pathogenesis of liver diseases or testing of drugs. Here, we developed two types of hydrogels by crosslinking hydrazide-functionalized gelatin with either oxidized dialdehyde hyaluronan or alginate through the formation of hydrazone bonds. Gel formulations were studied through texture analysis and rheometry, showing mechanical properties comparable to those of liver tissue while also demonstrating long-term stability. The biocompatibility of hydrogels and their ability to host hepatocytes was studied in vitro in comparison to pure gelatin hydrogels crosslinked by transglutaminase using the hepatocellular line HepG2. It was found that HepG2 cells could be successfully embedded in the hydrogels, showing no signs of gel toxicity and proliferating in a 3D environment comparable to pure transglutaminase cross-linked gelatin hydrogels used as control. Altogether, hydrazide gelatin in combination with oxidized polysaccharides makes stable in situ gelling systems for the incorporation of hepatocytes, which may pave the way for use in liver tissue engineering and drug testing.
Collapse
Affiliation(s)
- Christian Willems
- Department of Biomedical Materials, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, 06120 Halle, Germany; (C.W.); (F.Q.)
| | - Fangdi Qi
- Department of Biomedical Materials, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, 06120 Halle, Germany; (C.W.); (F.Q.)
| | - Marie-Luise Trutschel
- Department of Pharmaceutical Technology, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Thomas Groth
- Department of Biomedical Materials, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, 06120 Halle, Germany; (C.W.); (F.Q.)
- Interdisciplinary Center of Materials Science, Martin-Luther University Halle-Wittenberg, 06120 Halle, Germany
| |
Collapse
|
19
|
Bider F, Miola M, Clejanu CE, Götzelmann J, Kuth S, Vernè E, Basu B, Boccaccini AR. 3D bioprinting of multifunctional alginate dialdehyde (ADA)-gelatin (GEL) (ADA-GEL) hydrogels incorporating ferulic acid. Int J Biol Macromol 2024; 257:128449. [PMID: 38029911 DOI: 10.1016/j.ijbiomac.2023.128449] [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: 07/11/2023] [Revised: 10/17/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023]
Abstract
The present work explores the 3D extrusion printing of ferulic acid (FA)-containing alginate dialdehyde (ADA)-gelatin (GEL) scaffolds with a wide spectrum of biophysical and pharmacological properties. The tailored addition of FA (≤0.2 %) increases the crosslinking between FA and GEL in the presence of calcium chloride (CaCl2) and microbial transglutaminase, as confirmed using trinitrobenzenesulfonic acid (TNBS) assay. In agreement with an increase in crosslinking density, a higher viscosity of ADA-GEL with FA incorporation was achieved, leading to better printability. Importantly, FA release, enzymatic degradation and swelling were progressively reduced with an increase in FA loading to ADA-GEL, over 28 days. Similar positive impact on antibacterial properties with S. epidermidis strains as well as antioxidant properties were recorded. Intriguingly, FA incorporated ADA-GEL supported murine pre-osteoblast proliferation with reduced osteosarcoma cell proliferation over 7 days in culture, implicating potential anticancer property. Most importantly, FA-incorporated and cell-encapsulated ADA-GEL can be extrusion printed to shape fidelity-compliant multilayer scaffolds, which also support pre-osteoblast cells over 7 days in culture. Taken together, the present study has confirmed the significant potential of 3D bioprinting of ADA-GEL-FA ink to obtain structurally stable scaffolds with a broad spectrum of biophysical and therapeutically significant properties, for bone tissue engineering applications.
Collapse
Affiliation(s)
- Faina Bider
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Marta Miola
- Applied Science and Technology Department, Politecnico di Torino, 10129 Torino, Italy
| | - Corina-Elena Clejanu
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Johanna Götzelmann
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Sonja Kuth
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Enrica Vernè
- Applied Science and Technology Department, Politecnico di Torino, 10129 Torino, Italy
| | - Bikramjit Basu
- Materials Research Center, Indian Institute of Science Bangalore, Bangalore 560012, India
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, 91058 Erlangen, Germany.
| |
Collapse
|
20
|
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
|
21
|
Seymour AJ, Kilian D, Navarro RS, Hull SM, Heilshorn SC. 3D printing microporous scaffolds from modular bioinks containing sacrificial, cell-encapsulating microgels. Biomater Sci 2023; 11:7598-7615. [PMID: 37824082 PMCID: PMC10842430 DOI: 10.1039/d3bm00721a] [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] [Indexed: 10/13/2023]
Abstract
Microgel-based biomaterials have inherent porosity and are often extrudable, making them well-suited for 3D bioprinting applications. Cells are commonly introduced into these granular inks post-printing using cell infiltration. However, due to slow cell migration speeds, this strategy struggles to achieve depth-independent cell distributions within thick 3D printed geometries. To address this, we leverage granular ink modularity by combining two microgels with distinct functions: (1) structural, UV-crosslinkable microgels made from gelatin methacryloyl (GelMA) and (2) sacrificial, cell-laden microgels made from oxidized alginate (AlgOx). We hypothesize that encapsulating cells within sacrificial AlgOx microgels would enable the simultaneous introduction of void space and release of cells at depths unachievable through cell infiltration alone. Blending the microgels in different ratios produces a family of highly printable GelMA : AlgOx microgel inks with void fractions ranging from 0.03 to 0.35. As expected, void fraction influences the morphology of human umbilical vein endothelial cells (HUVEC) within GelMA : AlgOx inks. Crucially, void fraction does not alter the ideal HUVEC distribution seen throughout the depth of 3D printed samples. This work presents a strategy for fabricating constructs with tunable porosity and depth-independent cell distribution, highlighting the promise of microgel-based inks for 3D bioprinting.
Collapse
Affiliation(s)
- Alexis J Seymour
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - David Kilian
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA.
| | - Renato S Navarro
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA.
| | - Sarah M Hull
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Shahriari-Khalaji M, Sattar M, Cao R, Zhu M. Angiogenesis, hemocompatibility and bactericidal effect of bioactive natural polymer-based bilayer adhesive skin substitute for infected burned wound healing. Bioact Mater 2023; 29:177-195. [PMID: 37520303 PMCID: PMC10384635 DOI: 10.1016/j.bioactmat.2023.07.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023] Open
Abstract
Thermal wounds are complex and lethal with irregular shapes, risk of infection, slow healing, and large surface area. The mortality rate in patients with infected burns is twice that of non-infected burns. Developing multifunctional skin substitutes to augment the healing rate of infected burns is vital. Herein, we 3D printed a hydrogel scaffold comprising carboxymethyl chitosan (CMCs) and oxidized alginate grafted catechol (O-AlgCat) on a hydrophobic electrospun layer, forming a bilayer skin substitute (BSS). The functional layer (FL) was fabricated by physiochemical crosslinking to ensure favorable biodegradability. The gallium-containing hydrophobic electrospun layer or backing layer (BL) could mimic the epidermis of skin, avoiding fluid penetration and offering antibacterial activity. 3D printed FL contains catechol, gallium, and biologically active platelet rich fibrin (PRF) to adhere to both tissue and BL, show antibacterial activity, encourage angiogenesis, cell growth, and migration. The fabricated bioactive BSS exhibited noticeable adhesive properties (P ≤ 0.05), significant antibacterial activity (P ≤ 0.05), faster clot formation, and the potential to promote proliferation (P ≤ 0.05) and migration (P ≤ 0.05) of L929 cells. Furthermore, the angiogenesis was significantly higher (P ≤ 0.05) when evaluated in vivo and in ovo. The BSS-covered wounds healed faster due to low inflammation and high collagen density. Based on the obtained results, the fabricated bioactive BSS could be an effective treatment for infected burn wounds.
Collapse
Affiliation(s)
- Mina Shahriari-Khalaji
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Mamoona Sattar
- Research Group of Microbiological Engineering and Medical Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Ran Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, China
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
23
|
Shen KH, Chiu TH, Teng KC, Yu J, Yeh YC. Fabrication of triple-crosslinked gelatin/alginate hydrogels for controlled release applications. Int J Biol Macromol 2023; 250:126133. [PMID: 37543263 DOI: 10.1016/j.ijbiomac.2023.126133] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
Hydrogels have been demonstrated as smart drug carriers to recognize the tumor microenvironment for cancer treatment, where the dynamic crosslinks in the hydrogel network contribute to the stimuli-responsive features but also result in poor stability and weak mechanical property of the hydrogels. Here, phenylboronic acid-grafted polyethyleneimine (PBA-PEI)-modified gelatin (PPG) was synthesized to crosslink alginate dialdehyde (ADA) through imine bonds and boronate ester bonds, and then calcium ions (Ca2+) were added to introduce the third calcium-carboxylate crosslinking in the network to form the triple-crosslinked PPG/ADA-Ca2+ hydrogels. Given the three types of dynamic bonds in the network, PPG/ADA-Ca2+ hydrogels possessed a self-healing manner, stimuli-responsiveness, and better mechanical properties compared to single- or double-crosslinked hydrogels. The controlled release capability of PPG/ADA-Ca2+ hydrogels was also demonstrated, showing the encapsulated molecules can be rapidly released from the hydrogel network in the presence of hydrogen peroxide while the release rate can be slowed down at acidic pH. Furthermore, PPG/ADA-Ca2+ hydrogels presented selected cytotoxicity and drug delivery to cancer cells due to the regulated degradation by the cellular microenvironment. Taken together, PPG/ADA-Ca2+ hydrogels have been demonstrated as promising biomaterials with multiple desirable properties and dynamic features to perform controlled molecule release for biomedical applications.
Collapse
Affiliation(s)
- Ke-Han Shen
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Ting-Hsiang Chiu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Kuang-Chih Teng
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Jiashing Yu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Yi-Cheun Yeh
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
24
|
Kim NG, Kim SC, Kim TH, Je JY, Lee B, Lee SG, Kim YM, Kang HW, Qian ZJ, Kim N, Jung WK. Ishophloroglucin A-based multifunctional oxidized alginate/gelatin hydrogel for accelerating wound healing. Int J Biol Macromol 2023; 245:125484. [PMID: 37348579 DOI: 10.1016/j.ijbiomac.2023.125484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/12/2023] [Accepted: 06/17/2023] [Indexed: 06/24/2023]
Abstract
This study investigated the potential applicability of wound dressing hydrogels for tissue engineering, focusing on their ability to deliver pharmacological agents and absorb exudates. Specifically, we explored the use of polyphenols, as they have shown promise as bioactive and cross-linking agents in hydrogel fabrication. Ishophloroglucin A (IPA), a polyphenol not previously utilized in tissue engineering, was incorporated as both a drug and cross-linking agent within the hydrogel. We integrated the extracted IPA, obtained through the utilization of separation and purification techniques such as high-performance liquid chromatography (HPLC), liquid chromatography-mass spectrometry (LC-MS), and nuclear magnetic resonance (NMR) into oxidized alginate (OA) and gelatin (GEL) hydrogels. Our findings revealed that the mechanical properties, thermal stability, swelling, and degradation of the multifunctional hydrogel can be modulated via intermolecular interactions between the natural polymer and IPA. Moreover, the controlled release of IPA endows the hydrogel with antioxidant and antimicrobial characteristics. Overall, the wound healing efficacy, based on intermolecular interactions and drug potency, has been substantiated through accelerated wound closure and collagen deposition in an ICR mouse full-thickness wound model. These results suggest that incorporating IPA into natural polymers as both a drug and cross-linking agent has significant implications for tissue engineering applications.
Collapse
Affiliation(s)
- Nam-Gyun Kim
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-Senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
| | - Se-Chang Kim
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-Senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
| | - Tae-Hee Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Jae-Young Je
- Major of Human Bioconvergence, School of Smart Healthcare, Pukyong National University, Busan 48513, South Korea
| | - Bonggi Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Sang Gil Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea; Department of Smart Green Technology Engineering, Pukyong National University, Busan, 48513, South Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Hyun Wook Kang
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-Senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Zhong-Ji Qian
- College of Food Science and Technology, School of Chemistry and Environment, Shenzhen Institute of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Ocean University, Zhanjiang 524088, China; Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518114, Guangdong, China
| | - Namwon Kim
- Ingram School of Engineering, Texas State University, San Marcos, TX 78666, USA; Research Institute of Advanced Materials (RIAM), Seoul National University, Seoul 08826, Republic of Korea; Materials Science, Engineering, and Commercialization (MSEC), Texas State University, San Marcos, TX 78666, USA
| | - Won-Kyo Jung
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-Senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
25
|
Ritschl L, Schilling P, Wittmer A, Bohner M, Bernstein A, Schmal H, Seidenstuecker M. Composite material consisting of microporous beta-TCP ceramic and alginate-dialdehyde-gelatin for controlled dual release of clindamycin and bone morphogenetic protein 2. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:39. [PMID: 37498466 PMCID: PMC10374674 DOI: 10.1007/s10856-023-06743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
The aim of this study was to produce a composite of microporous β-TCP filled with alginate-gelatin crosslinked hydrogel, clindamycin and bone morphogenetic protein (BMP-2) to prolong the drug-release behaviour for up to 28 days. The most promising alginate-di-aldehyde(ADA)-gelatin gel for drug release from microcapsules was used to fill microporous β-TCP ceramics under directional flow in a special loading chamber. Dual release of clindamycin and BMP-2 was measured on days 1, 2, 3, 6, 9, 14, 21 and 28 by high performance liquid chromatography (HPLC) and enzyme-linked immunosorbent assay (ELISA). After release, the microbial efficacy of the clindamycin was checked and the biocompatibility of the composite was tested in cell culture. Clindamycin and the model substance FITC-protein A were released from microcapsules over 28 days. The clindamycin burst release was 43 ± 1%. For the loaded ceramics, a clindamycin release above the minimal inhibitory concentration (MIC) until day 9 and a burst release of 90.56 ± 2.96% were detected. BMP-2 was released from the loaded ceramics in low concentrations over 28 days. The release of active substances from β-TCP and hydrogel have already been extensively studied. Directional flow loading is a special procedure in which the ceramic could act as a stabilizer in the bone and, as a biodegradable system, enables a single-stage surgical procedure. Whether ADA-gelatin gel is suitable for this procedure as a more biodegradable alternative to pure alginate or whether a dual release is possible in this composite has not yet been investigated.
Collapse
Affiliation(s)
- Lucas Ritschl
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108, Freiburg, Germany.
| | - Pia Schilling
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108, Freiburg, Germany
| | - Annette Wittmer
- Medical Center Albert-Ludwigs-University of Freiburg, Institute of Microbiology and Hygiene, Hermann-Herder-Straße 11, 79104, Freiburg, Germany
| | - Marc Bohner
- Robert Mathys Foundation, Bischmattstrasse 12, 2544, Bettlach, Switzerland
| | - Anke Bernstein
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108, Freiburg, Germany
| | - Hagen Schmal
- Department of Orthopedics and Trauma Surgery, Medical Center Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Michael Seidenstuecker
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108, Freiburg, Germany.
| |
Collapse
|
26
|
Wang J, Liu S, Huang J, Ren K, Zhu Y, Yang S. Alginate: Microbial production, functionalization, and biomedical applications. Int J Biol Macromol 2023; 242:125048. [PMID: 37236570 DOI: 10.1016/j.ijbiomac.2023.125048] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/21/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
Alginates are natural polysaccharides widely participating in food, pharmaceutical, and environmental applications due to their excellent gelling capacity. Their excellent biocompatibility and biodegradability further extend their application to biomedical fields. The low consistency in molecular weight and composition of algae-based alginates may limit their performance in advanced biomedical applications. It makes microbial alginate production more attractive due to its potential for customizing alginate molecules with stable characteristics. Production costs remain the primary factor limiting the commercialization of microbial alginates. However, carbon-rich wastes from sugar, dairy, and biodiesel industries may serve as potential substitutes for pure sugars for microbial alginate production to reduce substrate costs. Fermentation parameter control and genetic engineering strategies may further improve the production efficiency and customize the molecular composition of microbial alginates. To meet the specific needs of biomedical applications, alginates may need functionalization, such as functional group modifications and crosslinking treatments, to achieve enhanced mechanical properties and biochemical activities. The development of alginate-based composites incorporated with other polysaccharides, gelatin, and bioactive factors can integrate the advantages of each component to meet multiple requirements in wound healing, drug delivery, and tissue engineering applications. This review provided a comprehensive insight into the sustainable production of high-value microbial alginates. It also discussed recent advances in alginate modification strategies and alginate-based composites for representative biomedical applications.
Collapse
Affiliation(s)
- Jianfei Wang
- Department of Chemical Engineering, SUNY College of Environmental Science and Forestry, Syracuse, NY 13210, United States
| | - Shijie Liu
- Department of Chemical Engineering, SUNY College of Environmental Science and Forestry, Syracuse, NY 13210, United States.
| | - Jiaqi Huang
- Department of Chemical Engineering, SUNY College of Environmental Science and Forestry, Syracuse, NY 13210, United States; The Center for Biotechnology & Interdisciplinary Studies (CBIS) at Rensselaer Polytechnic Institute, Troy, NY 12180, United States
| | - Kexin Ren
- Department of Chemical Engineering, SUNY College of Environmental Science and Forestry, Syracuse, NY 13210, United States
| | - Yan Zhu
- Department of Chemical Engineering, SUNY College of Environmental Science and Forestry, Syracuse, NY 13210, United States
| | - Siying Yang
- Department of Chemical Engineering, SUNY College of Environmental Science and Forestry, Syracuse, NY 13210, United States
| |
Collapse
|
27
|
Schulik J, Salehi S, Boccaccini AR, Schrüfer S, Schubert DW, Arkudas A, Kengelbach-Weigand A, Horch RE, Schmid R. Comparison of the Behavior of 3D-Printed Endothelial Cells in Different Bioinks. Bioengineering (Basel) 2023; 10:751. [PMID: 37508778 PMCID: PMC10376299 DOI: 10.3390/bioengineering10070751] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Biomaterials with characteristics similar to extracellular matrix and with suitable bioprinting properties are essential for vascular tissue engineering. In search for suitable biomaterials, this study investigated the three hydrogels alginate/hyaluronic acid/gelatin (Alg/HA/Gel), pre-crosslinked alginate di-aldehyde with gelatin (ADA-GEL), and gelatin methacryloyl (GelMA) with respect to their mechanical properties and to the survival, migration, and proliferation of human umbilical vein endothelial cells (HUVECs). In addition, the behavior of HUVECs was compared with their behavior in Matrigel. For this purpose, HUVECs were mixed with the inks both as single cells and as cell spheroids and printed using extrusion-based bioprinting. Good printability with shape fidelity was determined for all inks. The rheological measurements demonstrated the gelling consistency of the inks and shear-thinning behavior. Different Young's moduli of the hydrogels were determined. However, all measured values where within the range defined in the literature, leading to migration and sprouting, as well as reconciling migration with adhesion. Cell survival and proliferation in ADA-GEL and GelMA hydrogels were demonstrated for 14 days. In the Alg/HA/Gel bioink, cell death occurred within 7 days for single cells. Sprouting and migration of the HUVEC spheroids were observed in ADA-GEL and GelMA. Similar behavior of the spheroids was seen in Matrigel. In contrast, the spheroids in the Alg/HA/Gel ink died over the time studied. It has been shown that Alg/HA/Gel does not provide a good environment for long-term survival of HUVECs. In conclusion, ADA-GEL and GelMA are promising inks for vascular tissue engineering.
Collapse
Affiliation(s)
- Jana Schulik
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery University Hospital of Erlangen, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Sahar Salehi
- Chair of Biomaterials, University of Bayreuth, Prof.-Rüdiger-Bormann-Str. 1, 95447 Bayreuth, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstrasse 6, 91058 Erlangen, Germany
| | - Stefan Schrüfer
- Institute of Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Martensstraße 7, 91058 Erlangen, Germany
| | - Dirk W Schubert
- Institute of Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Martensstraße 7, 91058 Erlangen, Germany
| | - Andreas Arkudas
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery University Hospital of Erlangen, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Annika Kengelbach-Weigand
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery University Hospital of Erlangen, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Raymund E Horch
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery University Hospital of Erlangen, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Rafael Schmid
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery University Hospital of Erlangen, Krankenhausstraße 12, 91054 Erlangen, Germany
| |
Collapse
|
28
|
Karakaya E, Schöbel L, Zhong Y, Hazur J, Heid S, Forster L, Teßmar J, Boccaccini AR, Detsch R. How to Determine a Suitable Alginate for Biofabrication Approaches using an Extensive Alginate Library? Biomacromolecules 2023. [DOI: 10.1021/acs.biomac.2c01282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Affiliation(s)
- Emine Karakaya
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany
| | - Lisa Schöbel
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany
| | - Yu Zhong
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany
| | - Jonas Hazur
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany
| | - Susanne Heid
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany
| | - Leonard Forster
- Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Pleicherwall 2, Würzburg 97070, Germany
| | - Jörg Teßmar
- Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Pleicherwall 2, Würzburg 97070, Germany
| | - Aldo R. Boccaccini
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany
| | - Rainer Detsch
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany
| |
Collapse
|
29
|
Modification, 3D printing process and application of sodium alginate based hydrogels in soft tissue engineering: A review. Int J Biol Macromol 2023; 232:123450. [PMID: 36709808 DOI: 10.1016/j.ijbiomac.2023.123450] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/26/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
Sodium alginate (SA) is an inexpensive and biocompatible biomaterial with fast and gentle crosslinking that has been widely used in biological soft tissue repair/regeneration. Especially with the advent of 3D bioprinting technology, SA hydrogels have been applied more deeply in tissue engineering due to their excellent printability. Currently, the research on material modification, molding process and application of SA-based composite hydrogels has become a hot topic in tissue engineering, and a lot of fruitful results have been achieved. To better help readers have a comprehensive understanding of the development status of SA based hydrogels and their molding process in tissue engineering, in this review, we summarized SA modification methods, and provided a comparative analysis of the characteristics of various SA based hydrogels. Secondly, various molding methods of SA based hydrogels were introduced, the processing characteristics and the applications of different molding methods were analyzed and compared. Finally, the applications of SA based hydrogels in tissue engineering were reviewed, the challenges in their applications were also analyzed, and the future research directions were prospected. We believe this review is of great helpful for the researchers working in biomedical and tissue engineering.
Collapse
|
30
|
An injectable and pH-responsive hyaluronic acid hydrogel as metformin carrier for prevention of breast cancer recurrence. Carbohydr Polym 2023; 304:120493. [PMID: 36641175 DOI: 10.1016/j.carbpol.2022.120493] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022]
Abstract
To achieve the pH-responsive release of metformin in tumor acidic microenvironment, we prepared OHA-Met by covalently grafting metformin (Met) onto oxidized hyaluronic acid (OHA) through imine bonds, and then prepared carboxymethyl chitosan (CMCS)/OHA-Met drug loaded hydrogels. The CMCS/OHA-Met hydrogels showed the in-situ injection performance. At pH = 7.4, the cumulative release rate of metformin from CMCS/OHA-Met20 hydrogel was 42.7 ± 2.6 % in 6 h, and the release tended to balance after 72 h. At pH = 5.5, the release kept constant and the cumulative release rate was 79.3 ± 4.7 % at 6 h, showing good pH-responsive behavior. Metformin induced apoptosis of MCF-7 cells through the caspase 3/PARP pathway. CMCS/OHA-Met20 hydrogel could effectively kill MCF-7 cells, while reducing the cytotoxicity of free metformin to L929 cells. In vivo breast cancer recurrence experiments showed CMCS/OHA-Met20 hydrogel could achieve local injection and pH-responsive smart drug delivery at the tumor resection site, inhibiting breast cancer recurrence. Compared with direct administration, CMCS/OHA-Met20 hydrogel reduced the metformin dosage, frequency of administration and systemic side effects.
Collapse
|
31
|
Kara Özenler A, Distler T, Tihminlioglu F, Boccaccini AR. Fish scale containing alginate dialdehyde-gelatin bioink for bone tissue engineering. Biofabrication 2023; 15. [PMID: 36706451 DOI: 10.1088/1758-5090/acb6b7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/27/2023] [Indexed: 01/28/2023]
Abstract
The development of biomaterial inks suitable for biofabrication and mimicking the physicochemical properties of the extracellular matrix is essential for the application of bioprinting technology in tissue engineering (TE). The use of animal-derived proteinous materials, such as jellyfish collagen, or fish scale (FS) gelatin (GEL), has become an important pillar in biomaterial ink design to increase the bioactivity of hydrogels. However, besides the extraction of proteinous structures, the use of structurally intact FS as an additive could increase biocompatibility and bioactivity of hydrogels due to its organic (collagen) and inorganic (hydroxyapatite) contents, while simultaneously enhancing mechanical strength in three-dimensional (3D) printing applications. To test this hypothesis, we present here a composite biomaterial ink composed of FS and alginate dialdehyde (ADA)-GEL for 3D bioprinting applications. We fabricate 3D cell-laden hydrogels using mouse pre-osteoblast MC3T3-E1 cells. We evaluate the physicochemical and mechanical properties of FS incorporated ADA-GEL biomaterial inks as well as the bioactivity and cytocompatibility of cell-laden hydrogels. Due to the distinctive collagen orientation of the FS, the compressive strength of the hydrogels significantly increased with increasing FS particle content. Addition of FS also provided a tool to tune hydrogel stiffness. FS particles were homogeneously incorporated into the hydrogels. Particle-matrix integration was confirmed via scanning electron microscopy. FS incorporation in the ADA-GEL matrix increased the osteogenic differentiation of MC3T3-E1 cells in comparison to pristine ADA-GEL, as FS incorporation led to increased ALP activity and osteocalcin secretion of MC3T3-E1 cells. Due to the significantly increased stiffness and supported osteoinductivity of the hydrogels, FS structure as a natural collagen and hydroxyapatite source contributed to the biomaterial ink properties for bone engineering applications. Our findings indicate that ADA-GEL/FS represents a new biomaterial ink formulation with great potential for 3D bioprinting, and FS is confirmed as a promising additive for bone TE applications.
Collapse
Affiliation(s)
- Aylin Kara Özenler
- Department of Bioengineering, İzmir Institute of Technology, İzmir 35433, Turkey.,Institute of Biomaterials, Department of Material Science and Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen 91058, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden 01307, Germany
| | - Thomas Distler
- Institute of Biomaterials, Department of Material Science and Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen 91058, Germany
| | - Funda Tihminlioglu
- Department of Chemical Engineering, İzmir Institute of Technology, İzmir 35433, Turkey
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Material Science and Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen 91058, Germany
| |
Collapse
|
32
|
Weizel A, Distler T, Detsch R, Boccaccini AR, Seitz H, Budday S. Time-dependent hyper-viscoelastic parameter identification of human articular cartilage and substitute materials. J Mech Behav Biomed Mater 2023; 138:105618. [PMID: 36566662 DOI: 10.1016/j.jmbbm.2022.105618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022]
Abstract
Numerical simulations are a valuable tool to understand which processes during mechanical stimulations of hydrogels for cartilage replacement influence the behavior of chondrocytes and contribute to the success or failure of these materials as implants. Such simulations critically rely on the correct prediction of the material response through appropriate material models and corresponding parameters. In this study, we identify hyper-viscoelastic material parameters for numerical simulations in COMSOL Multiphysics® v. 5.6 for human articular cartilage and two replacement materials, the commercially available ChondroFillerliquid and oxidized alginate gelatin (ADA-GEL) hydrogels. We incorporate the realistic experimental boundary conditions into an inverse parameter identification scheme based on data from multiple loading modes simultaneously, including cyclic compression-tension and stress relaxation experiments. We provide individual parameter sets for the unconditioned and conditioned responses and discuss how viscoelastic effects are related to the materials' microstructure. ADA-GEL and ChondroFillerliquid exhibit faster stress relaxation than cartilage with lower relaxation time constants, while cartilage has the largest viscoelastic stress contribution. The elastic response predominates in ADA-GEL and ChondroFillerliquid, while the viscoelastic response predominates in cartilage. These results will help to simulate mechanical stimulations, support the development of suitable materials with distinct mechanical properties in the future and provide parameters and insight into the time-dependent material behavior of human articular cartilage.
Collapse
Affiliation(s)
- A Weizel
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Rostock, Germany.
| | - T Distler
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - R Detsch
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - A R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - H Seitz
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Rostock, Germany
| | - S Budday
- Institute of Applied Mechanics, Department of Mechanical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
33
|
Garshasbi HR, Naghib SM. Smart Stimuli-responsive Alginate Nanogels for Drug Delivery Systems and Cancer Therapy: A Review. Curr Pharm Des 2023; 29:3546-3562. [PMID: 38115614 DOI: 10.2174/0113816128283806231211073031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/13/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Nanogels are three-dimensional networks at the nanoscale level that can be fabricated through physical or chemical processes using polymers. These nanoparticles' biocompatibility, notable stability, efficacious drug-loading capacity, and ligand-binding proficiency make them highly suitable for employment as drug-delivery vehicles. In addition, they exhibit the ability to react to both endogenous and exogenous stimuli, which may include factors such as temperature, illumination, pH levels, and a diverse range of other factors. This facilitates the consistent administration of the drug to the intended site. Alginate biopolymers have been utilized to encapsulate anticancer drugs due to their biocompatible nature, hydrophilic properties, and cost-effectiveness. The efficacy of alginate nano gel-based systems in cancer treatment has been demonstrated through multiple studies that endorse their progress toward clinical implementation. This paper comprehensively reviews alginate and its associated systems in drug delivery systems.
Collapse
Affiliation(s)
- Hamid Reza Garshasbi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| |
Collapse
|
34
|
Antezana PE, Municoy S, Orive G, Desimone MF. Design of a New 3D Gelatin-Alginate Scaffold Loaded with Cannabis sativa Oil. Polymers (Basel) 2022; 14:4506. [PMID: 36365500 PMCID: PMC9658303 DOI: 10.3390/polym14214506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/15/2022] [Accepted: 10/21/2022] [Indexed: 09/20/2023] Open
Abstract
There is an increasing medical need for the development of new materials that could replace damaged organs, improve healing of critical wounds or provide the environment required for the formation of a new healthy tissue. The three-dimensional (3D) printing approach has emerged to overcome several of the major deficiencies of tissue engineering. The use of Cannabis sativa as a therapy for some diseases has spread throughout the world thanks to its benefits for patients. In this work, we developed a bioink made with gelatin and alginate that was able to be printed using an extrusion 3D bioprinter. The scaffolds obtained were lyophilized, characterized and the swelling was assessed. In addition, the scaffolds were loaded with Cannabis sativa oil extract. The presence of the extract provided antimicrobial and antioxidant activity to the 3D scaffolds. Altogether, our results suggest that the new biocompatible material printed with 3D technology and with the addition of Cannabis sativa oil could become an attractive alternative to common treatments of soft-tissue infections and wound repair.
Collapse
Affiliation(s)
- Pablo Edmundo Antezana
- Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
| | - Sofía Municoy
- Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01009 Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain
- University Institute for Regenerative Medicine and Oral Implantology-UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007 Vitoria-Gasteiz, Spain
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore
| | - Martín Federico Desimone
- Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
| |
Collapse
|
35
|
Ghorbani F, Kim M, Monavari M, Ghalandari B, Boccaccini AR. Mussel-inspired polydopamine decorated alginate dialdehyde-gelatin 3D printed scaffolds for bone tissue engineering application. Front Bioeng Biotechnol 2022; 10:940070. [PMID: 36003531 PMCID: PMC9393248 DOI: 10.3389/fbioe.2022.940070] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023] Open
Abstract
This study utilized extrusion-based 3D printing technology to fabricate calcium-cross-linked alginate dialdehyde-gelatin scaffolds for bone regeneration. The surface of polymeric constructs was modified with mussel-derived polydopamine (PDA) in order to induce biomineralization, increase hydrophilicity, and enhance cell interactions. Microscopic observations revealed that the PDA layer homogeneously coated the surface and did not appear to induce any distinct change in the microstructure of the scaffolds. The PDA-functionalized scaffolds were more mechanically stable (compression strength of 0.69 ± 0.02 MPa) and hydrophilic (contact angle of 26) than non-modified scaffolds. PDA-decorated ADA-GEL scaffolds demonstrated greater durability. As result of the 18-days immersion in simulated body fluid solution, the PDA-coated scaffolds showed satisfactory biomineralization. Based on theoretical energy analysis, it was shown that the scaffolds coated with PDA interact spontaneously with osteocalcin and osteomodulin (binding energy values of -35.95 kJ mol-1 and -46.39 kJ mol-1, respectively), resulting in the formation of a protein layer on the surface, suggesting applications in bone repair. PDA-coated ADA-GEL scaffolds are capable of supporting osteosarcoma MG-63 cell adhesion, viability (140.18% after 7 days), and proliferation. In addition to increased alkaline phosphatase secretion, osteoimage intensity also increased, indicating that the scaffolds could potentially induce bone regeneration. As a consequence, the present results confirm that 3D printed PDA-coated scaffolds constitute an intriguing novel approach for bone tissue engineering.
Collapse
Affiliation(s)
- Farnaz Ghorbani
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Minjoo Kim
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Mahshid Monavari
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Aldo R. Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
36
|
A thermo-sensitive hydrogel composed of methylcellulose/hyaluronic acid/silk fibrin as a biomimetic extracellular matrix to simulate breast cancer malignancy. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
37
|
Stagnoli S, Garro C, Ertekin O, Heid S, Seyferth S, Soria G, Mariano Correa N, Leal-Egaña A, Boccaccini AR. Topical Systems for the Controlled Release of Antineoplastic Drugs: Oxidized Alginate-Gelatin Hydrogel/Unilamellar Vesicles. J Colloid Interface Sci 2022; 629:1066-1080. [DOI: 10.1016/j.jcis.2022.08.163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 08/09/2022] [Accepted: 08/25/2022] [Indexed: 11/24/2022]
|
38
|
Varaprasad K, Karthikeyan C, Yallapu MM, Sadiku R. The significance of biomacromolecule alginate for the 3D printing of hydrogels for biomedical applications. Int J Biol Macromol 2022; 212:561-578. [DOI: 10.1016/j.ijbiomac.2022.05.157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/09/2022] [Accepted: 05/22/2022] [Indexed: 12/16/2022]
|
39
|
Schrade S, Ritschl L, Süss R, Schilling P, Seidenstuecker M. Gelatin Nanoparticles for Targeted Dual Drug Release out of Alginate-di-Aldehyde-Gelatin Gels. Gels 2022; 8:365. [PMID: 35735709 PMCID: PMC9222291 DOI: 10.3390/gels8060365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/19/2022] Open
Abstract
The aim of the present work was to develop a dual staged drug release of an antibiotic (clindamycin) and a growth factor: bone morphogenetic protein-2 (BMP-2) from a biodegradable system consisting of hydrogel and gelatin nanoparticles (GNP). Two-step de-solvation allowed us to prepare GNPs (~100 nm) as drug carriers. Fluorescein isothiocyanate (FITC)-conjugated protein A was used as a model substance for BMP-2. A 28-day release experiment was performed to determine the release kinetics from GNP for both FITC-protein A and BMP-2, and for clindamycin (CLI) from the hydrogel. The size, structure, and overall morphology of GNP samples (empty, loaded with FITC-protein A and BMP-2) were examined using an environmental scanning electron microscope (ESEM). Cell culture assays (Live/dead; cell proliferation; cytotoxicity) were performed with MG-63 cells and BMP-2-loaded GNPs. Drug release experiments using clindamycin-loaded alginate-di-aldehyde (ADA) gelatin gels containing the drug-loaded GNPs were performed for 28 days. The resulting GNPs showed an empty size of 117 ± 29 nm, 176 ± 15 nm and 216 ± 36 nm when containing 2% FITC-protein A and 1% BMP-2, respectively. No negative effects of BMP-2-loaded GNPs on MG-63 cells were observed in live/dead staining. In the proliferation assay, an increase in cell proliferation was observed for both GNPs (GNP + BMP-2 and controls). The cytotoxicity assay continuously showed very low cytotoxicity for GNPs (empty; loaded). Clindamycin release showed a concentration of 25-fold higher than the minimum inhibitory concentration (MIC) against Staphylococcus aureus throughout the 28 day period. BMP-2 showed a reduced burst release and a steady release (~2 µg/mL) over a 28 day period.
Collapse
Affiliation(s)
- Sophie Schrade
- G.E.R.N. Center of Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center—Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (S.S.); (L.R.); (P.S.)
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-University of Freiburg, Sonnenstr. 5, 79104 Freiburg, Germany;
| | - Lucas Ritschl
- G.E.R.N. Center of Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center—Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (S.S.); (L.R.); (P.S.)
| | - Regine Süss
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-University of Freiburg, Sonnenstr. 5, 79104 Freiburg, Germany;
| | - Pia Schilling
- G.E.R.N. Center of Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center—Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (S.S.); (L.R.); (P.S.)
| | - Michael Seidenstuecker
- G.E.R.N. Center of Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center—Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany; (S.S.); (L.R.); (P.S.)
| |
Collapse
|
40
|
Kara A, Distler T, Polley C, Schneidereit D, Seitz H, Friedrich O, Tihminlioglu F, Boccaccini AR. 3D printed gelatin/decellularized bone composite scaffolds for bone tissue engineering: Fabrication, characterization and cytocompatibility study. Mater Today Bio 2022; 15:100309. [PMID: 35757025 PMCID: PMC9213825 DOI: 10.1016/j.mtbio.2022.100309] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 11/18/2022] Open
Abstract
Three-dimensional (3D) printing technology enables the design of personalized scaffolds with tunable pore size and composition. Combining decellularization and 3D printing techniques provides the opportunity to fabricate scaffolds with high potential to mimic native tissue. The aim of this study is to produce novel decellularized bone extracellular matrix (dbECM)-reinforced composite-scaffold that can be used as a biomaterial for bone tissue engineering. Decellularized bone particles (dbPTs, ∼100 μm diameter) were obtained from rabbit femur and used as a reinforcement agent by mixing with gelatin (GEL) in different concentrations. 3D scaffolds were fabricated by using an extrusion-based bioprinter and crosslinking with microbial transglutaminase (mTG) enzyme, followed by freeze-drying to obtain porous structures. Fabricated 3D scaffolds were characterized morphologically, mechanically, and chemically. Furthermore, MC3T3-E1 mouse pre-osteoblast cells were seeded on the dbPTs reinforced GEL scaffolds (GEL/dbPTs) and cultured for 21 days to assess cytocompatibility and cell attachment. We demonstrate the 3D-printability of dbPTs-reinforced GEL hydrogels and the achievement of homogenous distribution of the dbPTs in the whole scaffold structure, as well as bioactivity and cytocompatibility of GEL/dbPTs scaffolds. It was shown that Young's modulus and degradation rate of scaffolds were enhanced with increasing dbPTs content. Multiphoton microscopy imaging displayed the interaction of cells with dbPTs, indicating attachment and proliferation of cells around the particles as well as into the GEL-particle hydrogels. Our results demonstrate that GEL/dbPTs hydrogel formulations have potential for bone tissue engineering.
Collapse
Affiliation(s)
- Aylin Kara
- İzmir Institute of Technology, Department of Bioengineering, İzmir, 35433, Turkey
- Institute of Biomaterials, Department of Material Science and Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91058, Germany
| | - Thomas Distler
- Institute of Biomaterials, Department of Material Science and Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91058, Germany
| | - Christian Polley
- Microfluidics, Department of Mechanical Engineering, University of Rostock, Rostock, 18059, Germany
| | - Dominik Schneidereit
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91052, Germany
| | - Hermann Seitz
- Microfluidics, Department of Mechanical Engineering, University of Rostock, Rostock, 18059, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91052, Germany
| | - Funda Tihminlioglu
- İzmir Institute of Technology, Department of Chemical Engineering, İzmir, 35433, Turkey
| | - Aldo R. Boccaccini
- Institute of Biomaterials, Department of Material Science and Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, 91058, Germany
| |
Collapse
|
41
|
|
42
|
Hyperelastic parameter identification of human articular cartilage and substitute materials. J Mech Behav Biomed Mater 2022; 133:105292. [DOI: 10.1016/j.jmbbm.2022.105292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 11/19/2022]
|
43
|
Kuth S, Karakaya E, Reiter N, Schmidt L, Paulsen F, Teßmar J, Budday S, Boccaccini AR. Oxidized Hyaluronic Acid-Gelatin-Based Hydrogels for Tissue Engineering and Soft Tissue Mimicking. Tissue Eng Part C Methods 2022; 28:301-313. [PMID: 35216525 DOI: 10.1089/ten.tec.2022.0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Hydrogels are ideal materials for mimicking and engineering soft tissue. Hyaluronic acid is a linear polysaccharide native to the human extracellular matrix. In this study, we first develop and characterize two hydrogel compositions built from oxidized HA and gelatin with and without alginate-di-aldehyde (ADA) crosslinked by ionic and enzymatic agents with potential applications in soft tissue engineering and tissue mimicking structures. The stability under incubation conditions was improved by adjusting crosslinking times. Through large-strain mechanical measurements, the hydrogels' properties were compared to human brain tissue and the samples containing ADA revealed similar mechanical properties to the native tissue specimens in cyclic compression-tension. In vitro characterization demonstrated a high viability of encapsulated mouse embryonic fibroblasts and a spreading of the cells in case of ADA-free samples.
Collapse
Affiliation(s)
- Sonja Kuth
- Institute of Biomaterials, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Emine Karakaya
- Institute of Biomaterials, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Nina Reiter
- Institute of Applied Mechanics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Laura Schmidt
- Institute of Biomaterials, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jörg Teßmar
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, Germany
| | - Silvia Budday
- Institute of Applied Mechanics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
44
|
Enzyme-Responsive Hydrogels as Potential Drug Delivery Systems-State of Knowledge and Future Prospects. Int J Mol Sci 2022; 23:ijms23084421. [PMID: 35457239 PMCID: PMC9031066 DOI: 10.3390/ijms23084421] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 12/25/2022] Open
Abstract
Fast advances in polymer science have provided new hydrogels for applications in drug delivery. Among modern drug formulations, polymeric type stimuli-responsive hydrogels (SRHs), also called smart hydrogels, deserve special attention as they revealed to be a promising tool useful for a variety of pharmaceutical and biomedical applications. In fact, the basic feature of these systems is the ability to change their mechanical properties, swelling ability, hydrophilicity, or bioactive molecules permeability, which are influenced by various stimuli, particularly enzymes. Indeed, among a great number of SHRs, enzyme-responsive hydrogels (ERHs) gain much interest as they possess several potential biomedical applications (e.g., in controlled release, drug delivery, etc.). Such a new type of SHRs directly respond to many different enzymes even under mild conditions. Therefore, they show either reversible or irreversible enzyme-induced changes both in chemical and physical properties. This article reviews the state-of-the art in ERHs designed for controlled drug delivery systems (DDSs). Principal enzymes used for biomedical hydrogel preparation were presented and different ERHs were further characterized focusing mainly on glucose oxidase-, β-galactosidase- and metalloproteinases-based catalyzed reactions. Additionally, strategies employed to produce ERHs were described. The current state of knowledge and the discussion were made on successful applications and prospects for further development of effective methods used to obtain ERH as DDSs.
Collapse
|
45
|
Jena SR, Dalei G, Das S, Nayak J, Pradhan M, Samanta L. Harnessing the potential of dialdehyde alginate-xanthan gum hydrogels as niche bioscaffolds for tissue engineering. Int J Biol Macromol 2022; 207:493-506. [PMID: 35276297 DOI: 10.1016/j.ijbiomac.2022.03.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/27/2022] [Accepted: 03/05/2022] [Indexed: 12/26/2022]
Abstract
Biomimetic hydrogels composed of natural polysaccharides have invariably blossomed as niche biomaterials in tissue engineering applications. The prospects of creating an extracellular matrix (ECM)-like milieu from such hydrogels has garnered considerable importance. In this study, we have fabricated bioscaffolds comprising dialdehyde alginate and xanthan gum and explored their potential use in tissue regeneration. The fabricated scaffolds displayed an interconnected porous network structure that is highly desirable for the aforesaid application. The scaffolds were endowed with good mechanical properties, thermostability, protein adsorption efficacy and degradability. Curcumin-loaded hydrogels exhibited appreciable antibacterial activity against E. coli. In vitro cytocompatibility studies revealed that the scaffolds promoted adhesion and proliferation of 3T3 fibroblast cells. The Western blot analysis of p53 gene indicated no growth arrest or apoptosis in 3T3 cells thus, signifying the non-toxic nature of the scaffolds. Furthermore, the ECM formation was confirmed via SDS-PAGE analysis. The overall results clearly validated these scaffolds as effectual biomaterials for tissue engineering applications.
Collapse
Affiliation(s)
- Soumya Ranjan Jena
- Redox Biology & Proteomics Laboratory, Department of Zoology and Centre of Excellence in Environment and Public Health, Ravenshaw University, Cuttack 753003, Odisha, India
| | - Ganeswar Dalei
- Department of Chemistry, Odisha University of Technology and Research, Bhubaneswar 751003, Odisha, India
| | - Subhraseema Das
- Department of Chemistry, Ravenshaw University, Cuttack 753003, Odisha, India.
| | - Jasmine Nayak
- Redox Biology & Proteomics Laboratory, Department of Zoology and Centre of Excellence in Environment and Public Health, Ravenshaw University, Cuttack 753003, Odisha, India
| | - Manoranjan Pradhan
- Department of Chemistry, Jhadeswar College of Engineering and Technology, Balasore 756056, Odisha, India
| | - Luna Samanta
- Redox Biology & Proteomics Laboratory, Department of Zoology and Centre of Excellence in Environment and Public Health, Ravenshaw University, Cuttack 753003, Odisha, India.
| |
Collapse
|
46
|
Zhu H, Monavari M, Zheng K, Distler T, Ouyang L, Heid S, Jin Z, He J, Li D, Boccaccini AR. 3D Bioprinting of Multifunctional Dynamic Nanocomposite Bioinks Incorporating Cu-Doped Mesoporous Bioactive Glass Nanoparticles for Bone Tissue Engineering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104996. [PMID: 35102718 DOI: 10.1002/smll.202104996] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/28/2021] [Indexed: 06/14/2023]
Abstract
Bioprinting has seen significant progress in recent years for the fabrication of bionic tissues with high complexity. However, it remains challenging to develop cell-laden bioinks exhibiting superior physiochemical properties and bio-functionality. In this study, a multifunctional nanocomposite bioink is developed based on amine-functionalized copper (Cu)-doped mesoporous bioactive glass nanoparticles (ACuMBGNs) and a hydrogel formulation relying on dynamic covalent chemistry composed of alginate dialdehyde (oxidized alginate) and gelatin, with favorable rheological properties, improved shape fidelity, and structural stability for extrusion-based bioprinting. The reversible dynamic microenvironment in combination with the impact of cell-adhesive ligands introduced by aminated particles enables the rapid spreading (within 3 days) and high survival (>90%) of embedded human osteosarcoma cells and immortalized mouse bone marrow-derived stroma cells. Osteogenic differentiation of primary mouse bone marrow stromal stem cells (BMSCs) and angiogenesis are promoted in the bioprinted alginate dialdehyde-gelatin (ADA-GEL or AG)-ACuMBGN scaffolds without additional growth factors in vitro, which is likely due to ion stimulation from the incorporated nanoparticles and possibly due to cell mechanosensing in the dynamic matrix. In conclusion, it is envisioned that these nanocomposite bioinks can serve as promising platforms for bioprinting complex 3D matrix environments providing superior physiochemical and biological performance for bone tissue engineering.
Collapse
Affiliation(s)
- Hui Zhu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Mahshid Monavari
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Kai Zheng
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Thomas Distler
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Liliang Ouyang
- Department of Mechanical Engineering, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Tsinghua University, Beijing, 100084, P. R. China
| | - Susanne Heid
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Zhaorui Jin
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
| | - Dichen Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
| | - Aldo R Boccaccini
- Department of Materials Science and Engineering Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nuremberg, 91058, Erlangen, Germany
| |
Collapse
|
47
|
Xie Y, Gao P, He F, Zhang C. Application of Alginate-Based Hydrogels in Hemostasis. Gels 2022; 8:109. [PMID: 35200490 PMCID: PMC8871293 DOI: 10.3390/gels8020109] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 12/24/2022] Open
Abstract
Hemorrhage, as a common trauma injury and clinical postoperative complication, may cause serious damage to the body, especially for patients with huge blood loss and coagulation dysfunction. Timely and effective hemostasis and avoidance of bleeding are of great significance for reducing body damage and improving the survival rate and quality of life of patients. Alginate is considered to be an excellent hemostatic polymer-based biomaterial due to its excellent biocompatibility, biodegradability, non-toxicity, non-immunogenicity, easy gelation and easy availability. In recent years, alginate hydrogels have been more and more widely used in the medical field, and a series of hemostatic related products have been developed such as medical dressings, hemostatic needles, transcatheter interventional embolization preparations, microneedles, injectable hydrogels, and hemostatic powders. The development and application prospects are extremely broad. This manuscript reviews the structure, properties and history of alginate, as well as the research progress of alginate hydrogels in clinical applications related to hemostasis. This review also discusses the current limitations and possible future development prospects of alginate hydrogels in hemostatic applications.
Collapse
Affiliation(s)
| | | | | | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Y.X.); (P.G.); (F.H.)
| |
Collapse
|
48
|
Khoshnood N, Zamanian A. Development of novel alginate‐polyethyleneimine cell‐laden bioink designed for 3D bioprinting of cutaneous wound healing scaffolds. J Appl Polym Sci 2022. [DOI: 10.1002/app.52227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Negin Khoshnood
- Biomaterials Research Group, Nanotechnology and Advanced Materials Department Materials and Energy Research Center (MERC) Tehran Iran
| | - Ali Zamanian
- Biomaterials Research Group, Nanotechnology and Advanced Materials Department Materials and Energy Research Center (MERC) Tehran Iran
| |
Collapse
|
49
|
Barbosa MAG, Xavier CPR, Pereira RF, Petrikaitė V, Vasconcelos MH. 3D Cell Culture Models as Recapitulators of the Tumor Microenvironment for the Screening of Anti-Cancer Drugs. Cancers (Basel) 2021; 14:190. [PMID: 35008353 PMCID: PMC8749977 DOI: 10.3390/cancers14010190] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Today, innovative three-dimensional (3D) cell culture models have been proposed as viable and biomimetic alternatives for initial drug screening, allowing the improvement of the efficiency of drug development. These models are gaining popularity, given their ability to reproduce key aspects of the tumor microenvironment, concerning the 3D tumor architecture as well as the interactions of tumor cells with the extracellular matrix and surrounding non-tumor cells. The development of accurate 3D models may become beneficial to decrease the use of laboratory animals in scientific research, in accordance with the European Union's regulation on the 3R rule (Replacement, Reduction, Refinement). This review focuses on the impact of 3D cell culture models on cancer research, discussing their advantages, limitations, and compatibility with high-throughput screenings and automated systems. An insight is also given on the adequacy of the available readouts for the interpretation of the data obtained from the 3D cell culture models. Importantly, we also emphasize the need for the incorporation of additional and complementary microenvironment elements on the design of 3D cell culture models, towards improved predictive value of drug efficacy.
Collapse
Affiliation(s)
- Mélanie A. G. Barbosa
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Cristina P. R. Xavier
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Rúben F. Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Biofabrication Group, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickevičiaus g 9, LT-44307 Kaunas, Lithuania;
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| | - M. Helena Vasconcelos
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
50
|
Rosiak P, Latanska I, Paul P, Sujka W, Kolesinska B. Modification of Alginates to Modulate Their Physic-Chemical Properties and Obtain Biomaterials with Different Functional Properties. Molecules 2021; 26:7264. [PMID: 34885846 PMCID: PMC8659150 DOI: 10.3390/molecules26237264] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 01/02/2023] Open
Abstract
Modified alginates have a wide range of applications, including in the manufacture of dressings and scaffolds used for regenerative medicine, in systems for selective drug delivery, and as hydrogel materials. This literature review discusses the methods used to modify alginates and obtain materials with new or improved functional properties. It discusses the diverse biological and functional activity of alginates. It presents methods of modification that utilize both natural and synthetic peptides, and describes their influence on the biological properties of the alginates. The success of functionalization depends on the reaction conditions being sufficient to guarantee the desired transformations and provide modified alginates with new desirable properties, but mild enough to prevent degradation of the alginates. This review is a literature description of efficient methods of alginate functionalization using biologically active ligands. Particular attention was paid to methods of alginate functionalization with peptides, because the combination of the properties of alginates and peptides leads to the obtaining of conjugates with properties resulting from both components as well as a completely new, different functionality.
Collapse
Affiliation(s)
- Piotr Rosiak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| | - Ilona Latanska
- Tricomed S.A., Swietojanska 5/9, 93-493 Lodz, Poland; (I.L.); (W.S.)
| | - Paulina Paul
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| | - Witold Sujka
- Tricomed S.A., Swietojanska 5/9, 93-493 Lodz, Poland; (I.L.); (W.S.)
| | - Beata Kolesinska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| |
Collapse
|