1
|
Zhao Y, Cao G, Wang Z, Liu D, Ren L, Ma D. The recent progress of bone regeneration materials containing EGCG. J Mater Chem B 2024; 12:9835-9844. [PMID: 39257355 DOI: 10.1039/d4tb00604f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Epigallocatechin-3-gallate (EGCG) is the most effective active ingredient in tea polyphenols and belongs to the category of catechins. EGCG has excellent antioxidant activity, anti-inflammatory, osteogenesis-promoting, and antibacterial properties, and has been widely studied in orthopedic diseases such as osteoporosis. To reach the lesion site, achieve sustained release, promote osteogenesis, regulate macrophage polarization, and improve the physical properties of materials, EGCG needs to be cross-linked or incorporated in bone regeneration materials. This article reviews the application of bone regeneration materials combined with EGCG, including natural polymer bone regeneration materials, synthetic polymer bone regeneration materials, bioceramic bone regeneration materials, metal bone regeneration materials, hydrogel bone regeneration materials and metal-EGCG networks. In addition, the fabrication methods for the regenerated scaffolds are also elaborated in the text. To sum up, it reveals the excellent development potential of materials containing EGCG and the shortcomings of current research, which will provide important reference for the future exploration of bone regeneration materials containing EGCG.
Collapse
Affiliation(s)
- Yaoye Zhao
- Lanzhou University Second Hospital, Lanzhou University, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou 730030, Gansu, China.
| | - Guoding Cao
- Lanzhou University Second Hospital, Lanzhou University, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou 730030, Gansu, China.
| | - Zixin Wang
- School of Stomatology, Lanzhou University, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou 730000, Gansu, China
| | - Desheng Liu
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, Gansu, China
| | - Liling Ren
- School of Stomatology, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Dongyang Ma
- Lanzhou University Second Hospital, Lanzhou University, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou 730030, Gansu, China.
| |
Collapse
|
2
|
Parvin N, Kumar V, Joo SW, Mandal TK. Cutting-Edge Hydrogel Technologies in Tissue Engineering and Biosensing: An Updated Review. MATERIALS (BASEL, SWITZERLAND) 2024; 17:4792. [PMID: 39410363 PMCID: PMC11477805 DOI: 10.3390/ma17194792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/22/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024]
Abstract
Hydrogels, known for their unique ability to retain large amounts of water, have emerged as pivotal materials in both tissue engineering and biosensing applications. This review provides an updated and comprehensive examination of cutting-edge hydrogel technologies and their multifaceted roles in these fields. Initially, the chemical composition and intrinsic properties of both natural and synthetic hydrogels are discussed, highlighting their biocompatibility and biodegradability. The manuscript then probes into innovative scaffold designs and fabrication techniques such as 3D printing, electrospinning, and self-assembly methods, emphasizing their applications in regenerating bone, cartilage, skin, and neural tissues. In the realm of biosensing, hydrogels' responsive nature is explored through their integration into optical, electrochemical, and piezoelectric sensors. These sensors are instrumental in medical diagnostics for glucose monitoring, pathogen detection, and biomarker identification, as well as in environmental and industrial applications like pollution and food quality monitoring. Furthermore, the review explores cross-disciplinary innovations, including the use of hydrogels in wearable devices, and hybrid systems, and their potential in personalized medicine. By addressing current challenges and future directions, this review aims to underscore the transformative impact of hydrogel technologies in advancing healthcare and industrial practices, thereby providing a vital resource for researchers and practitioners in the field.
Collapse
Affiliation(s)
| | | | - Sang Woo Joo
- School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea; (N.P.); (V.K.)
| | - Tapas Kumar Mandal
- School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea; (N.P.); (V.K.)
| |
Collapse
|
3
|
Staples R, Ivanovski S, Vaswani K, Vaquette C. Melt electrowriting scaffolds with fibre-guiding features for periodontal attachment. Acta Biomater 2024; 180:337-357. [PMID: 38583749 DOI: 10.1016/j.actbio.2024.04.006] [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: 10/10/2023] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/09/2024]
Abstract
Periodontal regeneration requires the re-attachment of oblique and perpendicular periodontal ligament (PDL) fibres to newly formed cementum and alveolar bone, which has proven elusive with existing approaches. In this study, multiple fibre-guiding biphasic tissue engineered constructs were fabricated by melt electrowriting. The biphasic scaffolds were 95 % porous and consisted of a pore size gradient bone compartment and periodontal compartment made of fibre-guiding channels with micro-architectural features ranging from 100 to 60 µm aimed to direct PDL fibre alignment and attachment. In vitro evaluations over 3 and 7 days demonstrated a marked improvement in collagen fibre orientation (over 60 % fully aligned) for scaffolds with micro-architecture ≤100 µm. The biphasic scaffolds were placed on a dentine slice and implanted ectopically, and this demonstrated that all micro-channels groups facilitated oblique and perpendicular alignment and attachment on the dentine with a mean nuclei angle and mean collagen fibre angle of approximately 60° resembling the native periodontal ligament attachment. A further in vivo testing using a surgically created rodent periodontal model highlighted the 80 µm micro-channel group's effectiveness, showing a significant increase in oblique PDL fibre attachment (72 %) and periodontal regeneration (56 %) when compared to all other groups onto the tooth root compared to control groups. Further to this, immunohistochemistry demonstrated the presence of periostin in the newly formed ligament indicating that functional regeneration occurred These findings suggest that scaffold micro-architectures of 100 µm or below can play a crucial role in directing periodontal tissue regeneration, potentially addressing a critical gap in periodontal therapy. STATEMENT OF SIGNIFICANCE: Periodontal regeneration remains a significant clinical challenge. Essential to restoring dental health and function is the proper attachment of the periodontal ligament, which is functionally oriented, to regenerated bone and cementum. Our research presents an innovative biphasic scaffold, utilizing Melt Electrowriting to systematically guide tissue growth. Distinct from existing methods, our scaffold is highly porous, adaptable, and precisely guides periodontal ligament fibre attachment to the opposing tooth root and alveolar bone interfaces, a critical step for achieving periodontal functional regeneration. Our findings not only bridge a significant gap in biomaterial driven tissue guidance but also promise more predictable outcomes for patients, marking a transformative advancement in the field.
Collapse
Affiliation(s)
- Reuben Staples
- The University of Queensland, School of Dentistry, Herston 4006, QLD, Australia; Centre for Orofacial Regeneration Reconstruction and Rehabilitation (COR3), Herston, QLD 4006, Australia
| | - Sašo Ivanovski
- The University of Queensland, School of Dentistry, Herston 4006, QLD, Australia; Centre for Orofacial Regeneration Reconstruction and Rehabilitation (COR3), Herston, QLD 4006, Australia
| | - Kanchan Vaswani
- The University of Queensland, School of Dentistry, Herston 4006, QLD, Australia; Centre for Orofacial Regeneration Reconstruction and Rehabilitation (COR3), Herston, QLD 4006, Australia
| | - Cedryck Vaquette
- The University of Queensland, School of Dentistry, Herston 4006, QLD, Australia; Centre for Orofacial Regeneration Reconstruction and Rehabilitation (COR3), Herston, QLD 4006, Australia.
| |
Collapse
|
4
|
Ozkendir O, Karaca I, Cullu S, Erdoğan OC, Yaşar HN, Dikici S, Owen R, Aldemir Dikici B. Engineering periodontal tissue interfaces using multiphasic scaffolds and membranes for guided bone and tissue regeneration. BIOMATERIALS ADVANCES 2024; 157:213732. [PMID: 38134730 DOI: 10.1016/j.bioadv.2023.213732] [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: 10/22/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Periodontal diseases are one of the greatest healthcare burdens worldwide. The periodontal tissue compartment is an anatomical tissue interface formed from the periodontal ligament, gingiva, cementum, and bone. This multifaceted composition makes tissue engineering strategies challenging to develop due to the interface of hard and soft tissues requiring multiphase scaffolds to recreate the native tissue architecture. Multilayer constructs can better mimic tissue interfaces due to the individually tuneable layers. They have different characteristics in each layer, with modulation of mechanical properties, material type, porosity, pore size, morphology, degradation properties, and drug-releasing profile all possible. The greatest challenge of multilayer constructs is to mechanically integrate consecutive layers to avoid delamination, especially when using multiple manufacturing processes. Here, we review the development of multilayer scaffolds that aim to recapitulate native periodontal tissue interfaces in terms of physical, chemical, and biological characteristics. Important properties of multiphasic biodegradable scaffolds are highlighted and summarised, with design requirements, biomaterials, and fabrication methods, as well as post-treatment and drug/growth factor incorporation discussed.
Collapse
Affiliation(s)
- Ozgu Ozkendir
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Ilayda Karaca
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Selin Cullu
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Oğul Can Erdoğan
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Hüsniye Nur Yaşar
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Serkan Dikici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey
| | - Robert Owen
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Betül Aldemir Dikici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir 35433, Turkey.
| |
Collapse
|
5
|
Tang Y, Yang X, Hu H, Jiang H, Xiong W, Mei H, Hu Y. Elevating the potential of CAR-T cell therapy in solid tumors: exploiting biomaterials-based delivery techniques. Front Bioeng Biotechnol 2024; 11:1320807. [PMID: 38312512 PMCID: PMC10835794 DOI: 10.3389/fbioe.2023.1320807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/05/2023] [Indexed: 02/06/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cells exhibit promising progress in addressing hematologic malignancies. However, CAR-T therapy for solid tumors remains limited, with no FDA-approved CAR-T products available for clinical use at present. Primary reasons include insufficient infiltration, accumulation, tumor immunosuppression of the microenvironment, and related side effects. Single utilization of CAR-T cannot effectively overcome these unfavorable obstacles. A probable effective pathway to achieve a better CAR-T therapy effect would be to combine the benefits of biomaterials-based technology. In this article, comprehensive biomaterials strategies to break through these obstacles of CAR-T cell therapy at the tumor sites are summarized, encompassing the following aspects: 1) generating orthotopic CAR-T cells; 2) facilitating CAR-T cell trafficking; 3) stimulating CAR-T cell expansion and infiltration; 4) improving CAR-T cell activity and persistence; 5) reprogramming the immunosuppressive microenvironments. Additionally, future requirements for the development of this field, with a specific emphasis on promoting innovation and facilitating clinical translation, are thoroughly discussed.
Collapse
Affiliation(s)
- Yuxiang Tang
- Tongji Medical College, Union Hospital, Institute of Hematology, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Xiaoyu Yang
- Department of Pharmacy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Hu
- School of Pharmacy, ChangZhou University, Changzhou, China
| | - Huiwen Jiang
- Tongji Medical College, Union Hospital, Institute of Hematology, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Wei Xiong
- Wuhan Sian Medical Technology Co., Ltd., Wuhan, China
| | - Heng Mei
- Tongji Medical College, Union Hospital, Institute of Hematology, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Yu Hu
- Tongji Medical College, Union Hospital, Institute of Hematology, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| |
Collapse
|
6
|
Ghalia MA, Alhanish A. Mechanical and biodegradability of porous PCL/PEG copolymer-reinforced cellulose nanofibers for soft tissue engineering applications. Med Eng Phys 2023; 120:104055. [PMID: 37838404 DOI: 10.1016/j.medengphy.2023.104055] [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: 05/16/2023] [Revised: 08/21/2023] [Accepted: 09/18/2023] [Indexed: 10/16/2023]
Abstract
The design and development of a new class of biomaterial has gained particular interest in producing polymer scaffold for biomedical applications. Mechanical properties, biological and controlling pores scaffold of the biomaterials are important factors to encourage cell growth and eventual tissue repair and regeneration. In this study, poly-ε-caprolactone (PCL) /polyethylene glycol (PEG) copolymer (80/20) incorporated with CNF scaffolds were made employing solvent casting and particulate leaching methods. Four mass percentages of CNF (1, 2.5, 5, and 10 wt.%) were integrated into the copolymer through a silane coupling agent. Mechanical properties were determined using Tensile Tester data acquisition to investigate the effect of porosity, pore size, and CNF contents. Tensile strength obtained for PCL/PEG- 5 wt.% CNF was 16 MPa, which drastically decreased after creating a porous structure to 7.1 MPa. The optimum parameters of the results were found to be 5 wt.% for CNF, 240 μm for pore size, and 83% for porosity. Scanning electron microscopy (SEM) micrograph reveals that consistent pore size and regular pore shape were accomplished after the addition of CNF-5 wt.% into PCL/PEG. The results of mass loss of PCL/PEG reinforced-CNF 1 % have clearly enhanced to double values compared with PCL/PEG copolymer and three times with PCL/PEG scaffold-CNF 1 %. In addition, all PCL/PEG reinforced and scaffold- CNF were partially disintegrated under composting conditions confirming their biodegradable behavior. This also provides a possible solution for the end life of these biomaterials.
Collapse
Affiliation(s)
| | - Atika Alhanish
- Department of Chemical Engineering, Faculty Oil, Gas and Renewable Energy Engineering, University of Zawia, Libya
| |
Collapse
|
7
|
Hu B, Gao J, Lu Y, Wang Y. Applications of Degradable Hydrogels in Novel Approaches to Disease Treatment and New Modes of Drug Delivery. Pharmaceutics 2023; 15:2370. [PMID: 37896132 PMCID: PMC10610366 DOI: 10.3390/pharmaceutics15102370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 10/29/2023] Open
Abstract
Hydrogels are particularly suitable materials for loading drug delivery agents; their high water content provides a biocompatible environment for most biomolecules, and their cross-linked nature protects the loaded agents from damage. During delivery, the delivered substance usually needs to be released gradually over time, which can be achieved by degradable cross-linked chains. In recent years, biodegradable hydrogels have become a promising technology in new methods of disease treatment and drug delivery methods due to their many advantageous properties. This review briefly discusses the degradation mechanisms of different types of biodegradable hydrogel systems and introduces the specific applications of degradable hydrogels in several new methods of disease treatment and drug delivery methods.
Collapse
Affiliation(s)
- Bo Hu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (B.H.); (J.G.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Jinyuan Gao
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (B.H.); (J.G.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Yu Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (B.H.); (J.G.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Yuji Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (B.H.); (J.G.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
8
|
Bourdon L, Attik N, Belkessam L, Chevalier C, Bousige C, Brioude A, Salles V. Direct-Writing Electrospun Functionalized Scaffolds for Periodontal Regeneration: In Vitro Studies. J Funct Biomater 2023; 14:jfb14050263. [PMID: 37233373 DOI: 10.3390/jfb14050263] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
Multiphasic scaffolds that combine different architectural, physical, and biological properties are the best option for the regeneration of complex tissues such as the periodontium. Current developed scaffolds generally lack architectural accuracy and rely on multistep manufacturing, which is difficult to implement for clinical applications. In this context, direct-writing electrospinning (DWE) represents a promising and rapid technique for developing thin 3D scaffolds with controlled architecture. The current study aimed to elaborate a biphasic scaffold using DWE based on two polycaprolactone solutions with interesting properties for bone and cement regeneration. One of the two scaffold parts contained hydroxyapatite nanoparticles (HAP) and the other contained the cementum protein 1 (CEMP1). After morphological characterizations, the elaborated scaffolds were assessed regarding periodontal ligament (PDL) cells in terms of cell proliferation, colonization, and mineralization ability. The results demonstrated that both HAP- and CEMP1-functionalized scaffolds were colonized by PDL cells and enhanced mineralization ability compared to unfunctionalized scaffolds, as revealed by alizarin red staining and OPN protein fluorescent expression. Taken together, the current data highlighted the potential of functional and organized scaffolds to stimulate bone and cementum regeneration. Moreover, DWE could be used to develop smart scaffolds with the ability to spatially control cellular orientation with suitable cellular activity at the micrometer scale, thereby enhancing periodontal and other complex tissue regeneration.
Collapse
Affiliation(s)
- Laura Bourdon
- Laboratoire des Multimatériaux et Interfaces, UMR 5615, CNRS, Université Claude Bernard Lyon 1, Bâtiment Chevreul, 6 Rue Victor Grignard, 69622 Villeurbanne, France
| | - Nina Attik
- Laboratoire des Multimatériaux et Interfaces, UMR 5615, CNRS, Université Claude Bernard Lyon 1, Bâtiment Chevreul, 6 Rue Victor Grignard, 69622 Villeurbanne, France
- Faculté d'Odontologie, Université Lyon 1, 11 Rue Guillaume Paradin, 69008 Lyon, France
| | - Liza Belkessam
- Laboratoire des Multimatériaux et Interfaces, UMR 5615, CNRS, Université Claude Bernard Lyon 1, Bâtiment Chevreul, 6 Rue Victor Grignard, 69622 Villeurbanne, France
- Faculté d'Odontologie, Université Lyon 1, 11 Rue Guillaume Paradin, 69008 Lyon, France
| | - Charlène Chevalier
- Laboratoire des Multimatériaux et Interfaces, UMR 5615, CNRS, Université Claude Bernard Lyon 1, Bâtiment Chevreul, 6 Rue Victor Grignard, 69622 Villeurbanne, France
- Faculté d'Odontologie, Université Lyon 1, 11 Rue Guillaume Paradin, 69008 Lyon, France
| | - Colin Bousige
- Laboratoire des Multimatériaux et Interfaces, UMR 5615, CNRS, Université Claude Bernard Lyon 1, Bâtiment Chevreul, 6 Rue Victor Grignard, 69622 Villeurbanne, France
| | - Arnaud Brioude
- Laboratoire des Multimatériaux et Interfaces, UMR 5615, CNRS, Université Claude Bernard Lyon 1, Bâtiment Chevreul, 6 Rue Victor Grignard, 69622 Villeurbanne, France
| | - Vincent Salles
- Laboratoire des Multimatériaux et Interfaces, UMR 5615, CNRS, Université Claude Bernard Lyon 1, Bâtiment Chevreul, 6 Rue Victor Grignard, 69622 Villeurbanne, France
- LIMMS, CNRS-IIS UMI 2820, The University of Tokyo, Tokyo 153-8505, Japan
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan
| |
Collapse
|
9
|
Atia GAN, Shalaby HK, Ali NG, Morsy SM, Ghobashy MM, Attia HAN, Barai P, Nady N, Kodous AS, Barai HR. New Challenges and Prospective Applications of Three-Dimensional Bioactive Polymeric Hydrogels in Oral and Craniofacial Tissue Engineering: A Narrative Review. Pharmaceuticals (Basel) 2023; 16:702. [PMID: 37242485 PMCID: PMC10224377 DOI: 10.3390/ph16050702] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Regenerative medicine, and dentistry offers enormous potential for enhancing treatment results and has been fueled by bioengineering breakthroughs over the previous few decades. Bioengineered tissues and constructing functional structures capable of healing, maintaining, and regenerating damaged tissues and organs have had a broad influence on medicine and dentistry. Approaches for combining bioinspired materials, cells, and therapeutic chemicals are critical in stimulating tissue regeneration or as medicinal systems. Because of its capacity to maintain an unique 3D form, offer physical stability for the cells in produced tissues, and replicate the native tissues, hydrogels have been utilized as one of the most frequent tissue engineering scaffolds during the last twenty years. Hydrogels' high water content can provide an excellent conditions for cell viability as well as an architecture that mimics real tissues, bone, and cartilage. Hydrogels have been used to enable cell immobilization and growth factor application. This paper summarizes the features, structure, synthesis and production methods, uses, new challenges, and future prospects of bioactive polymeric hydrogels in dental and osseous tissue engineering of clinical, exploring, systematical and scientific applications.
Collapse
Affiliation(s)
- Gamal Abdel Nasser Atia
- Department of Oral Medicine, Periodontology, and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia P.O. Box 41522, Egypt
| | - Hany K. Shalaby
- Department of Oral Medicine, Periodontology and Oral Diagnosis, Faculty of Dentistry, Suez University, Suez P.O. Box 43512, Egypt
| | - Naema Goda Ali
- Department of Oral Medicine, Periodontology, and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia P.O. Box 41522, Egypt
| | - Shaimaa Mohammed Morsy
- Department of Oral Medicine, Periodontology, and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia P.O. Box 41522, Egypt
| | - Mohamed Mohamady Ghobashy
- Radiation Research of Polymer Chemistry Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo P.O. Box 13759, Egypt
| | - Hager Abdel Nasser Attia
- Department of Molecular Biology and Chemistry, Faculty of Science, Alexandria University, Alexandria P.O. Box 21526, Egypt
| | - Paritosh Barai
- Department of Biochemistry and Molecular Biology, Primeasia University, Dhaka 1213, Bangladesh
| | - Norhan Nady
- Polymeric Materials Research Department, Advanced Technology and New Materials Research Institute (ATNMRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg Elarab, Alexandria P.O. Box 21934, Egypt
| | - Ahmad S. Kodous
- Department of Radiation Biology, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Cairo P.O. Box 13759, Egypt
| | - Hasi Rani Barai
- Department of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
10
|
Dal-Fabbro R, Swanson WB, Capalbo LC, Sasaki H, Bottino MC. Next-generation biomaterials for dental pulp tissue immunomodulation. Dent Mater 2023; 39:333-349. [PMID: 36894414 PMCID: PMC11034777 DOI: 10.1016/j.dental.2023.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVES The current standard for treating irreversibly damaged dental pulp is root canal therapy, which involves complete removal and debridement of the pulp space and filling with an inert biomaterial. A regenerative approach to treating diseased dental pulp may allow for complete healing of the native tooth structure and enhance the long-term outcome of once-necrotic teeth. The aim of this paper is, therefore, to highlight the current state of dental pulp tissue engineering and immunomodulatory biomaterials properties, identifying exciting opportunities for their synergy in developing next-generation biomaterials-driven technologies. METHODS An overview of the inflammatory process focusing on immune responses of the dental pulp, followed by periapical and periodontal tissue inflammation are elaborated. Then, the most recent advances in treating infection-induced inflammatory oral diseases, focusing on biocompatible materials with immunomodulatory properties are discussed. Of note, we highlight some of the most used modifications in biomaterials' surface, or content/drug incorporation focused on immunomodulation based on an extensive literature search over the last decade. RESULTS We provide the readers with a critical summary of recent advances in immunomodulation related to pulpal, periapical, and periodontal diseases while bringing light to tissue engineering strategies focusing on healing and regenerating multiple tissue types. SIGNIFICANCE Significant advances have been made in developing biomaterials that take advantage of the host's immune system to guide a specific regenerative outcome. Biomaterials that efficiently and predictably modulate cells in the dental pulp complex hold significant clinical promise for improving standards of care compared to endodontic root canal therapy.
Collapse
Affiliation(s)
- Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - W Benton Swanson
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Leticia C Capalbo
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Hajime Sasaki
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
11
|
Sugiaman VK, Jeffrey, Naliani S, Pranata N, Djuanda R, Saputri RI. Polymeric Scaffolds Used in Dental Pulp Regeneration by Tissue Engineering Approach. Polymers (Basel) 2023; 15:1082. [PMID: 36904323 PMCID: PMC10007583 DOI: 10.3390/polym15051082] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023] Open
Abstract
Currently, the challenge in dentistry is to revitalize dental pulp by utilizing tissue engineering technology; thus, a biomaterial is needed to facilitate the process. One of the three essential elements in tissue engineering technology is a scaffold. A scaffold acts as a three-dimensional (3D) framework that provides structural and biological support and creates a good environment for cell activation, communication between cells, and inducing cell organization. Therefore, the selection of a scaffold represents a challenge in regenerative endodontics. A scaffold must be safe, biodegradable, and biocompatible, with low immunogenicity, and must be able to support cell growth. Moreover, it must be supported by adequate scaffold characteristics, which include the level of porosity, pore size, and interconnectivity; these factors ultimately play an essential role in cell behavior and tissue formation. The use of natural or synthetic polymer scaffolds with excellent mechanical properties, such as small pore size and a high surface-to-volume ratio, as a matrix in dental tissue engineering has recently received a lot of attention because it shows great potential with good biological characteristics for cell regeneration. This review describes the latest developments regarding the usage of natural or synthetic scaffold polymers that have the ideal biomaterial properties to facilitate tissue regeneration when combined with stem cells and growth factors in revitalizing dental pulp tissue. The utilization of polymer scaffolds in tissue engineering can help the pulp tissue regeneration process.
Collapse
Affiliation(s)
- Vinna K. Sugiaman
- Department of Oral Biology, Faculty of Dentistry, Maranatha Christian University, Bandung 40164, West Java, Indonesia
| | - Jeffrey
- Department of Pediatric Dentistry, Faculty of Dentistry, Jenderal Achmad Yani University, Cimahi 40531, West Java, Indonesia
| | - Silvia Naliani
- Department of Prosthodontics, Faculty of Dentistry, Maranatha Christian University, Bandung 40164, West Java, Indonesia
| | - Natallia Pranata
- Department of Oral Biology, Faculty of Dentistry, Maranatha Christian University, Bandung 40164, West Java, Indonesia
| | - Rudy Djuanda
- Department of Conservative Dentistry and Endodontic, Faculty of Dentistry, Maranatha Christian University, Bandung 40164, West Java, Indonesia
| | - Rosalina Intan Saputri
- College of Medicine, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Faculty of Dentistry, Maranatha Christian University, Bandung 40164, West Java, Indonesia
| |
Collapse
|
12
|
Yamamoto T, Randriantsilefisoa R, Sprecher CM, D’Este M. Fabrication of Collagen-Hyaluronic Acid Cryogels by Directional Freezing Mimicking Cartilage Arcade-like Structure. Biomolecules 2022; 12:biom12121809. [PMID: 36551237 PMCID: PMC9775592 DOI: 10.3390/biom12121809] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/23/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
The internal architecture of tissue-like constructs is fundamental to their structural and biological functions. Here, we introduce a simple and robust method to fabricate cryogels based on derivatized extracellular matrix (ECM) macromolecules with porosity arranged according to the typical Benninghoff zonal architecture of articular cartilage. To obtain this arcade-like structure, the technique used the growth of ice crystals from copper pins at cryogenic temperatures. The directional cryogel formation enabled the organized growth of ice crystals over a large distance (>4 mm). The compositional properties were achieved by forming double networks (DNs) of hyaluronic acid and collagen derivatives (MeHA and CollGTA, respectively), which also served to improve the mechanical properties of the otherwise weak collagen scaffolds. Compositionally biomimetic and more resilient MeHA-CollGTA DNs (Young's modulus ≈ 200 kilopascals) were therefore produced. The technique presented expands the fabrication methods available for providing ECM macromolecules with architectural elements mimicking cartilage complexity.
Collapse
|
13
|
Bhushan S, Singh S, Maiti TK, Sharma C, Dutt D, Sharma S, Li C, Tag Eldin EM. Scaffold Fabrication Techniques of Biomaterials for Bone Tissue Engineering: A Critical Review. Bioengineering (Basel) 2022; 9:728. [PMID: 36550933 PMCID: PMC9774188 DOI: 10.3390/bioengineering9120728] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/27/2022] Open
Abstract
Bone tissue engineering (BTE) is a promising alternative to repair bone defects using biomaterial scaffolds, cells, and growth factors to attain satisfactory outcomes. This review targets the fabrication of bone scaffolds, such as the conventional and electrohydrodynamic techniques, for the treatment of bone defects as an alternative to autograft, allograft, and xenograft sources. Additionally, the modern approaches to fabricating bone constructs by additive manufacturing, injection molding, microsphere-based sintering, and 4D printing techniques, providing a favorable environment for bone regeneration, function, and viability, are thoroughly discussed. The polymers used, fabrication methods, advantages, and limitations in bone tissue engineering application are also emphasized. This review also provides a future outlook regarding the potential of BTE as well as its possibilities in clinical trials.
Collapse
Affiliation(s)
- Sakchi Bhushan
- Department of Paper Technology, IIT Roorkee, Saharanpur 247001, India
| | - Sandhya Singh
- Department of Paper Technology, IIT Roorkee, Saharanpur 247001, India
| | - Tushar Kanti Maiti
- Department of Polymer and Process Engineering, IIT Roorkee, Saharanpur 247001, India
| | - Chhavi Sharma
- Department of Polymer and Process Engineering, IIT Roorkee, Saharanpur 247001, India
| | - Dharm Dutt
- Department of Paper Technology, IIT Roorkee, Saharanpur 247001, India
| | - Shubham Sharma
- Mechanical Engineering Department, University Center for Research & Development, Chandigarh University, Mohali 140413, India
- School of Mechanical and Automotive Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Changhe Li
- School of Mechanical and Automotive Engineering, Qingdao University of Technology, Qingdao 266520, China
| | | |
Collapse
|
14
|
Upadhyay U, Sireesha S, Gupta S, Sreedhar I, Anitha K. Freeze v/s air-dried alginate-pectin gel beads modified with sodium dodecyl sulphate for enhanced removal of copper ions. Carbohydr Polym 2022; 301:120294. [DOI: 10.1016/j.carbpol.2022.120294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022]
|
15
|
Li M, Song P, Wang W, Xu Y, Li J, Wu L, Gui X, Zeng Z, Zhou Z, Liu M, Kong Q, Fan Y, Zhang X, Zhou C, Liu L. Preparation and characterization of biomimetic gradient multi-layer cell-laden scaffolds for osteochondral integrated repair. J Mater Chem B 2022; 10:4172-4188. [PMID: 35531933 DOI: 10.1039/d2tb00576j] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A cell-laden tissue engineering scaffold for osteochondral integrated repair is one of the ideal strategies for osteochondral lesions. In this study, we fabricated cell-laden porous hydrogel scaffolds with gradient nano-hydroxyapatite using methacrylic anhydride gelatin (GelMA), nano-hydroxyapatite (nHA), and polyethylene oxide (PEO) solution for osteochondral tissue regeneration. The scaffold possessed interconnected pores and a nano-hydroxyapatite gradient in the vertical direction. The chemical, physical, mechanical, and biological properties of the hydrogel solutions and scaffolds were characterized. In vitro experiments confirmed that cells were distributed homogeneously and that different pore structures could affect the proliferation and differentiation of BMSCs. The Nonporous hydrogel was beneficial for the chondrogenic differentiation of BMSCs and interconnected pores were conducive to BMSC proliferation and osteogenic differentiation. The osteochondral integrative repair capacity of the scaffold was assessed by implanting the scaffolds into the intercondylar defect of the rabbit femur. By constructing pore structures in different layers, the cells in different layers of the hydrogels were in an intrinsic environment for survival and differentiation. Animal experiments confirmed that tissue engineering scaffolds for osteochondral lesions require different pore structures in different layers, and gradient structure facilitated integrated repair.
Collapse
Affiliation(s)
- Mingxin Li
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ping Song
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China. .,College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Wenzhao Wang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yang Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China. .,College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Jun Li
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Lina Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China. .,College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Xingyu Gui
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China. .,College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Zhimou Zeng
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Zhigang Zhou
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ming Liu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qingquan Kong
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China. .,College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China. .,College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China. .,College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Lei Liu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
16
|
Ye S, Wei B, Zeng L. Advances on Hydrogels for Oral Science Research. Gels 2022; 8:gels8050302. [PMID: 35621600 PMCID: PMC9140480 DOI: 10.3390/gels8050302] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 11/16/2022] Open
Abstract
Hydrogels are biocompatible polymer systems, which have become a hotspot in biomedical research. As hydrogels mimic the structure of natural extracellular matrices, they are considered as good scaffold materials in the tissue engineering area for repairing dental pulp and periodontal damages. Combined with different kinds of stem cells and growth factors, various hydrogel complexes have played an optimistic role in endodontic and periodontal tissue engineering studies. Further, hydrogels exhibit biological effects in response to external stimuli, which results in hydrogels having a promising application in local drug delivery. This review summarized the advances of hydrogels in oral science research, in the hopes of providing a reference for future applications.
Collapse
Affiliation(s)
- Shengjia Ye
- Department of Prosthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China;
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China
| | - Bin Wei
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China
- Department of Stomatology Special Consultation Clinic, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Correspondence: (B.W.); (L.Z.)
| | - Li Zeng
- Department of Prosthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China;
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China
- Correspondence: (B.W.); (L.Z.)
| |
Collapse
|
17
|
Tarrahi R, Khataee A, Karimi A, Yoon Y. The latest achievements in plant cellulose-based biomaterials for tissue engineering focusing on skin repair. CHEMOSPHERE 2022; 288:132529. [PMID: 34637866 DOI: 10.1016/j.chemosphere.2021.132529] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/27/2021] [Accepted: 10/08/2021] [Indexed: 06/13/2023]
Abstract
The present work reviews recent developments in plant cellulose-based biomaterial design and applications, properties, characterizations, and synthesis for skin tissue engineering and wound healing. Cellulose-based biomaterials are promising materials for their remarkable adaptability with three-dimensional polymeric structure. They are capable of mimicking tissue properties, which plays a key role in tissue engineering. Besides, concerns for environmental issues have motivated scientists to move toward eco-friendly materials and natural polymer-based materials for applications in the tissue engineering field these days. Therefore, cellulose as an appropriate substitute for common polymers based on crude coal, animal, and human-derived biomolecules is greatly considered for various applications in biomedical fields. Generally, natural biomaterials lack good mechanical properties for skin tissue engineering. But using modified cellulose-based biopolymers tackles these restrictions and prevents immunogenic responses. Moreover, tissue engineering is a quick promoting field focusing on the generation of novel biomaterials with modified characteristics to improve scaffold function through physical, biochemical, and chemical tailoring. Also, nanocellulose with a broad range of applications, particularly in tissue engineering, advanced wound dressing, and as a material for coupling with drugs and sensorics, has been reviewed here. Moreover, the potential cytotoxicity and immunogenicity of cellulose-based biomaterials are addressed in this review.
Collapse
Affiliation(s)
- Roshanak Tarrahi
- Health Promotion Research Center, Iran University of Medical Sciences, 14496-14535, Tehran, Iran
| | - Alireza Khataee
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, 51666-16471, Tabriz, Iran; Department of Environmental Engineering, Gebze Technical University, 41400, Gebze, Turkey
| | - Afzal Karimi
- Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, 1449614535, Tehran, Iran
| | - Yeojoon Yoon
- Department of Environmental and Energy Engineering, Yonsei University, Wonju, Republic of Korea
| |
Collapse
|
18
|
Demir D, Uğurlu MA, Ceylan S, Sakım B, Genç R, Bölgen N. Assessment of Chitosan‐Gum Tragacanth Cryogels For Tissue Engineering Applications. POLYM INT 2022. [DOI: 10.1002/pi.6372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Didem Demir
- Mersin University, Engineering Faculty, Chemical Engineering Department Mersin Turkey
| | - Müge Aşık Uğurlu
- Mersin University, Engineering Faculty, Chemical Engineering Department Mersin Turkey
| | - Seda Ceylan
- Adana Alparslan Türkeş Science and Technology University, Engineering Faculty, Bioengineering Department Adana Turkey
| | - Burcu Sakım
- Mersin University, Engineering Faculty, Chemical Engineering Department Mersin Turkey
| | - Rükan Genç
- Mersin University, Engineering Faculty, Chemical Engineering Department Mersin Turkey
| | - Nimet Bölgen
- Mersin University, Engineering Faculty, Chemical Engineering Department Mersin Turkey
| |
Collapse
|
19
|
Zhang Y, Zhang M, Cheng D, Xu S, Du C, Xie L, Zhao W. Applications of electrospun scaffolds with enlarged pores in tissue engineering. Biomater Sci 2022; 10:1423-1447. [DOI: 10.1039/d1bm01651b] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Despite electrospinning has multiple advantages over other methods such as creating materials with superfine fiber diameter, high specific surface area, and good mechanical properties, the pore diameter of scaffolds prepared...
Collapse
|
20
|
Rezaeeyazdi M, Colombani T, Eggermont LJ, Bencherif SA. Engineering hyaluronic acid-based cryogels for CD44-mediated breast tumor reconstruction. Mater Today Bio 2022; 13:100207. [PMID: 35198956 PMCID: PMC8844817 DOI: 10.1016/j.mtbio.2022.100207] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/19/2022] [Accepted: 01/22/2022] [Indexed: 01/03/2023] Open
Abstract
Breast cancer is a major health concern worldwide and is the leading cause of cancer-related death among American women. Traditional therapies, such as surgery, chemotherapy, and radiotherapy, are usually ineffective. Furthermore, cancer recurrence following targeted therapy often results from acquired drug resistance. Therefore, more realistic tumor models than monolayer cell culture for drug screening and discovery in an in vitro setting would facilitate the development of new therapeutic strategies. Toward this goal, we first developed a simple, rapid, low-cost, and high-throughput method for generating uniform multi-cellular tumor spheroids (MCTS) with controllable size. Next, biomimetic cryogel scaffolds fabricated from hyaluronic acid (HA) were utilized as a platform to reconstruct breast tumor microtissues with aspects of the complex tumor microenvironment in three dimensions. Finally, we investigated the interactions between the HA-based cryogels and CD44-positive breast tumor cells, individually or as MCTS. We found that incorporating the adhesive RGD peptide in cryogels led to the formation of a monolayer of tumor cells on the polymer walls, whereas MCTS cultured on RGD-free HA cryogels resulted in the growth of large and dense microtumors, more similar to native tumor masses. As a result, the MCTS-laden HA cryogel system induced a highly aggressive and chemotherapy drug-resistant tumor model. RGD-free HA-based cryogels represent an effective starting point for designing tumor models for preclinical research, therapeutic drug screening, and early cancer diagnosis.
Collapse
Affiliation(s)
| | - Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Loek J. Eggermont
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
21
|
Biocompatible and Biomaterials Application in Drug Delivery System in Oral Cavity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9011226. [PMID: 34812267 PMCID: PMC8605911 DOI: 10.1155/2021/9011226] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/27/2021] [Indexed: 02/03/2023]
Abstract
Biomaterials applications have rapidly expanded into different fields of sciences. One of the important fields of using biomaterials is dentistry, which can facilitate implantation, surgery, and treatment of oral diseases such as peri-implantitis, periodontitis, and other dental problems. Drug delivery systems based on biocompatible materials play a vital role in the release of drugs into aim tissues of the oral cavity with minimum side effects. Therefore, scientists have studied various delivery systems to improve the efficacy and acceptability of therapeutic approaches in dental problems and oral diseases. Also, biomaterials could be utilized as carriers in biocompatible drug delivery systems. For instance, natural polymeric substances, such as gelatin, chitosan, calcium phosphate, alginate, and xanthan gum are used to prepare different forms of delivery systems. In addition, some alloys are conducted in drug complexes for the better in transportation. Delivery systems based on biomaterials are provided with different strategies, although individual biomaterial has advantages and disadvantages which have a significant influence on transportation of complex such as solubility in physiological environments or distribution in tissues. Biomaterials have antibacterial and anti-inflammatory effects and prolonged time contact and even enhance antibiotic activities in oral infections. Moreover, these biomaterials are commonly prepared in some forms such as particulate complex, fibers, microspheres, gels, hydrogels, and injectable systems. In this review, we examined the application of biocompatible materials in drug delivery systems of oral and dental diseases or problems.
Collapse
|
22
|
Bayrak G, Perçin I, Kılıç Süloğlu A, Denizli A. Amino acid functionalized macroporous gelatin cryogels: Characterization and effects on cell proliferation. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
23
|
Chen Z, Xiao H, Zhang H, Xin Q, Zhang H, Liu H, Wu M, Zuo L, Luo J, Guo Q, Ding C, Tan H, Li J. Heterogenous hydrogel mimicking the osteochondral ECM applied to tissue regeneration. J Mater Chem B 2021; 9:8646-8658. [PMID: 34595487 DOI: 10.1039/d1tb00518a] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Inspired by the intricate extracellular matrix (ECM) of natural cartilage and subchondral bone, a heterogenous bilayer hydrogel scaffold is fabricated. Gelatin methacrylate (GelMA) and acryloyl glucosamine (AGA) serve as the main components in the upper layer, mimicking the chondral ECM. Meanwhile, vinylphosphonic acid (VPA) as a non-collagen protein analogue is incorporated into the bottom layer to induce the in situ biomineralization of calcium phosphate. The two heterogenous layers are effectively sutured together by the inter-diffusion between the upper and bottom layer hydrogels, together with chelation between the calcium ions and alginate added to separate layers. The interfacial bonding between the two different layers was thoroughly investigated via rheological measurements. The incorporation of AGA promotes chondrocytes to produce collagen type II and glycosaminoglycans and upregulates the expression of chondrogenesis-related genes. In addition, the minerals induced by VPA facilitate the osteogenesis of bone marrow mesenchymal stem cells (BMSCs). In vivo evaluation confirms the biocompatibility of the scaffold with minor inflammation and confirms the best repair ability of the bilayer hydrogel. This cell-free, cost-effective and efficient hydrogel shows great potential for osteochondral repair and inspires the design of other tissue-engineering scaffolds.
Collapse
Affiliation(s)
- Zhuoxin Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Hong Xiao
- Department of Pain Management, West China Hospital, Sichuan University, No. 37, GuoXue Xiang, Chengdu 610041, China
| | - Hongbo Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Qiangwei Xin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Haochen Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Haixin Liu
- Department of Orthopedics, People's Hospital of Deyang City, No. 173, Taishan North Road, Deyang 618000, China
| | - Mingzhen Wu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Liangrui Zuo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Jun Luo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Qiang Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chunmei Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China. .,CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Beijing 100190, China
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China. .,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.,Med-X Center for Materials, Sichuan University, 610041, China
| |
Collapse
|
24
|
Hosseini M, Shafiee A. Engineering Bioactive Scaffolds for Skin Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101384. [PMID: 34313003 DOI: 10.1002/smll.202101384] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Indexed: 06/13/2023]
Abstract
Large skin wounds pose a major clinical challenge. Scarcity of donor site and postsurgical scarring contribute to the incomplete or partial loss of function and aesthetic concerns in skin wound patients. Currently, a wide variety of skin grafts are being applied in clinical settings. Scaffolds are used to overcome the issues related to the misaligned architecture of the repaired skin tissues. The current review summarizes the contribution of biomaterials to wound healing and skin regeneration and addresses the existing limitations in skin grafting. Then, the clinically approved biologic and synthetic skin substitutes are extensively reviewed. Next, the techniques for modification of skin grafts aiming for enhanced tissue regeneration are outlined, and a summary of different growth factor delivery systems using biomaterials is presented. Considering the significant progress in biomaterial science and manufacturing technologies, the idea of biomaterial-based skin grafts with the ability for scarless wound healing and reconstructing full skin organ is more achievable than ever.
Collapse
Affiliation(s)
- Motaharesadat Hosseini
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD, 4059, Australia
| | - Abbas Shafiee
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| |
Collapse
|
25
|
Lv H, Wu B, Song J, Wu W, Cai W, Xu J. Hydrogel, a novel therapeutic and delivery strategy, in the treatment of intrauterine adhesions. J Mater Chem B 2021; 9:6536-6552. [PMID: 34324619 DOI: 10.1039/d1tb01005k] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Intrauterine adhesions (IUAs) are caused by damage to the underlying lining of the endometrium. They' re related to disorder of endometrial repair. In recent years, hydrogels with controllable biological activity have been widely used for treating IUAs. They encapsulate estrogen, cytokines, cells, or exosomes, forming a delivery system to release therapeutic components for the treatment of IUAs. In addition, the hydrogel acting as a barrier can be degraded in the body automatically, reducing the risk of infection caused by secondary surgeries. In this review, we summarize the recent progress of hydrogels and their application in IUAs as both a novel alternative therapeutic and an artificial delivery strategy.
Collapse
Affiliation(s)
- Houyi Lv
- Department of Reproductive Medicine, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | | | | | | | | | | |
Collapse
|
26
|
Lin CY, Kang JH. Mechanical Properties of Compact Bone Defined by the Stress-Strain Curve Measured Using Uniaxial Tensile Test: A Concise Review and Practical Guide. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4224. [PMID: 34361418 PMCID: PMC8347989 DOI: 10.3390/ma14154224] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/03/2022]
Abstract
Mechanical properties are crucial parameters for scaffold design for bone tissue engineering; therefore, it is important to understand the definitions of the mechanical properties of bones and relevant analysis methods, such that tissue engineers can use this information to properly design the mechanical properties of scaffolds for bone tissue engineering. The main purpose of this article is to provide a review and practical guide to understand and analyze the mechanical properties of compact bone that can be defined and extracted from the stress-strain curve measured using uniaxial tensile test until failure. The typical stress-strain curve of compact bone measured using uniaxial tensile test until failure is a bilinear, monotonically increasing curve. The associated mechanical properties can be obtained by analyzing this bilinear stress-strain curve. In this article, a computer programming code for analyzing the bilinear stress-strain curve of compact bone for quantifying the associated mechanical properties is provided, such that the readers can use this computer code to perform the analysis directly. In addition to being applied to compact bone, the information provided by this article can also be applied to quantify the mechanical properties of any material having a bilinear stress-strain curve, such as a whole bone, some metals and biomaterials. The information provided by this article can be applied by tissue engineers, such that they can have a reference to properly design the mechanical properties of scaffolds for bone tissue engineering. The information can also be applied by researchers in biomechanics and orthopedics to compare the mechanical properties of bones in different physiological or pathological conditions.
Collapse
Affiliation(s)
- Che-Yu Lin
- Institute of Applied Mechanics, College of Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Jiunn-Horng Kang
- Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, 252 Wuxing Str., Taipei 11031, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Str., Taipei 11031, Taiwan
- Research Center of Artificial Intelligence in Medicine, Taipei Medical University, 250 Wuxing Str., Taipei 11031, Taiwan
| |
Collapse
|
27
|
Boulais L, Jellali R, Pereira U, Leclerc E, Bencherif SA, Legallais C. Cryogel-Integrated Biochip for Liver Tissue Engineering. ACS APPLIED BIO MATERIALS 2021; 4:5617-5626. [PMID: 35006744 DOI: 10.1021/acsabm.1c00425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Microfluidic systems and polymer hydrogels have been widely developed for tissue engineering. Yet, only a few tools combining both approaches, especially for in vitro liver models, are being explored. In this study, an alginate-based cryogel-integrated biochip was engineered for dynamic hepatoma cell line culture in three dimensions (3D). The alginate cryogel was covalently cross-linked in the biochip at subzero temperatures (T < 0 °C) to create a scaffold with high mechanical stability and an interconnected macroporous network. By varying the alginate concentration and the cross-linker ratio, Young's modulus of the cryogel can be fine-tuned between 1.5 and 29 kPa, corresponding to the range of stiffness of the different physiological states of the liver. We demonstrated that HepG2/C3A cells can be cultured and maintained as viable under dynamic conditions in this device up to 6 days. Albumin synthesis and glucose consumption increased over the cell culture days. Moreover, a 3D cell structure was observed across the entire height of the biochip, which was preserved following alginate lyase treatment to remove the cryogel-based scaffold. In summary, these results represent a proof of concept of an interesting cell culture technology that should be further investigated to engineer healthy and cirrhotic liver models.
Collapse
Affiliation(s)
- Lilandra Boulais
- Université de Technologie de Compiègne, UMR CNRS 7338 Biomécanique et Bioingénierie, Centre de Recherche de Royallieu, Compiègne 60203, France
| | - Rachid Jellali
- Université de Technologie de Compiègne, UMR CNRS 7338 Biomécanique et Bioingénierie, Centre de Recherche de Royallieu, Compiègne 60203, France
| | - Ulysse Pereira
- Université de Technologie de Compiègne, UMR CNRS 7338 Biomécanique et Bioingénierie, Centre de Recherche de Royallieu, Compiègne 60203, France
| | - Eric Leclerc
- Université de Technologie de Compiègne, UMR CNRS 7338 Biomécanique et Bioingénierie, Centre de Recherche de Royallieu, Compiègne 60203, France
| | - Sidi A Bencherif
- Université de Technologie de Compiègne, UMR CNRS 7338 Biomécanique et Bioingénierie, Centre de Recherche de Royallieu, Compiègne 60203, France.,Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115-5005, United States.,Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115-5005, United States.,Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Cécile Legallais
- Université de Technologie de Compiègne, UMR CNRS 7338 Biomécanique et Bioingénierie, Centre de Recherche de Royallieu, Compiègne 60203, France
| |
Collapse
|
28
|
Mokhtari F, Azimi B, Salehi M, Hashemikia S, Danti S. Recent advances of polymer-based piezoelectric composites for biomedical applications. J Mech Behav Biomed Mater 2021; 122:104669. [PMID: 34280866 DOI: 10.1016/j.jmbbm.2021.104669] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/19/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023]
Abstract
Over the past decades, electronics have become central to many aspects of biomedicine and wearable device technologies as a promising personalized healthcare platform. Lead-free piezoelectric materials for converting mechanical into electrical energy through piezoelectric transduction are of significant value in a diverse range of technological applications. Organic piezoelectric biomaterials have attracted widespread attention as the functional materials in the biomedical devices due to their advantages of excellent biocompatibility. They include synthetic and biological polymers. Many biopolymers have been discovered to possess piezoelectricity in an appreciable amount, however their investigation is still preliminary. Due to their piezoelectric properties, better known synthetic fluorinated polymers have been intensively investigated and applied in biomedical applications including controlled drug delivery systems, tissue engineering, microfluidic and artificial muscle actuators, among others. Piezoelectric polymers, especially poly (vinylidene fluoride) (PVDF) and its copolymers are increasingly receiving interest as smart biomaterials due to their ability to convert physiological movements to electrical signals when in a controllable and reproducible manner. Despite possessing the greatest piezoelectric coefficients among all piezoelectric polymers, it is often desirable to increase the electrical outputs. The most promising routes toward significant improvements in the piezoelectric response and energy-harvesting performance of such materials is loading them with various inorganic nanofillers and/or applying some modification during the fabrication process. This paper offers a comprehensive review of the principles, properties, and applications of organic piezoelectric biomaterials (polymers and polymer/ceramic composites) with special attention on PVDF-based polymers and their composites in sensors, drug delivery and tissue engineering. Subsequently focuses on the most common fabrication routes to produce piezoelectric scaffolds, tissue and sensors which is electrospinning process. Promising upcoming strategies and new piezoelectric materials and fabrication techniques for these applications are presented to enable a future integration among these applications.
Collapse
Affiliation(s)
- Fatemeh Mokhtari
- Intelligent Polymer Research Institute, University of Wollongong, Wollongong NSW, Australia
| | - Bahareh Azimi
- Department of Civil and Industrial Engineering, University of Pisa, Pisa, Italy; Department. of Civil and Environmental Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Maryam Salehi
- Department of Civil Engineering, The University of Memphis, Memphis, TN, USA
| | - Samaneh Hashemikia
- Faculty of Textile Engineering, Urmia University of Technology, Urmia, Iran
| | - Serena Danti
- Department of Civil and Industrial Engineering, University of Pisa, Pisa, Italy; Department. of Civil and Environmental Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
| |
Collapse
|
29
|
Gsib O, Eggermont LJ, Egles C, Bencherif SA. Engineering a macroporous fibrin-based sequential interpenetrating polymer network for dermal tissue engineering. Biomater Sci 2021; 8:7106-7116. [PMID: 33089849 DOI: 10.1039/d0bm01161d] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The success of skin tissue engineering for deep wound healing relies predominantly on the design of innovative and effective biomaterials. This study reports the synthesis and characterization of a new type of naturally-derived and macroporous interpenetrating polymer network (IPN) for skin repair. These biomaterials consist of a biologically active fibrous fibrin network polymerized within a mechanically robust and macroporous construct made of polyethylene glycol and biodegradable serum albumin (PEGDM-co-SAM). First, mesoporous PEGDM-co-SAM hydrogels were synthesized and subjected to cryotreatment to introduce an interconnected macroporous network. Subsequently, fibrin precursors were incorporated within the cryotreated PEG-based network and then allowed to spontaneously polymerize and form a sequential IPN. Rheological measurements indicated that fibrin-based sequential IPN hydrogels exhibited improved and tunable mechanical properties when compared to fibrin hydrogels alone. In vitro data showed that human dermal fibroblasts adhere, infiltrate and proliferate within the IPN constructs, and were able to secrete endogenous extracellular matrix proteins, namely collagen I and fibronectin. Furthermore, a preclinical study in mice demonstrated that IPNs were stable over 1-month following subcutaneous implantation, induced a minimal host inflammatory response, and displayed a substantial cellular infiltration and tissue remodeling within the constructs. Collectively, these data suggest that macroporous and mechanically reinforced fibrin-based sequential IPN hydrogels are promising three-dimensional platforms for dermal tissue regeneration.
Collapse
Affiliation(s)
- Olfat Gsib
- Laboratoire de BioMécanique et BioIngénierie (BMBI), UMR CNRS 7388, Sorbonne Universités, Université de Technologie of Compiègne (UTC), Compiègne, France.
| | - Loek J Eggermont
- Departments of Chemical Engineering and Bioengineering, Northeastern University, Boston, MA, USA
| | - Christophe Egles
- Laboratoire de BioMécanique et BioIngénierie (BMBI), UMR CNRS 7388, Sorbonne Universités, Université de Technologie of Compiègne (UTC), Compiègne, France.
| | - Sidi A Bencherif
- Laboratoire de BioMécanique et BioIngénierie (BMBI), UMR CNRS 7388, Sorbonne Universités, Université de Technologie of Compiègne (UTC), Compiègne, France. and Departments of Chemical Engineering and Bioengineering, Northeastern University, Boston, MA, USA and Department of Bioengineering, Northeastern University, Boston, MA, USA and Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| |
Collapse
|
30
|
Müller D, Klepzig LF, Schlosser A, Dorfs D, Bigall NC. Structural Diversity in Cryoaerogel Synthesis. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:5109-5117. [PMID: 33884880 PMCID: PMC8154879 DOI: 10.1021/acs.langmuir.0c03619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Different techniques that enable the selective microstructure design of aerogels without the use of additives are presented. For this, aerogels were prepared from platinum nanoparticle solutions using the cryoaerogelation method, and respective impacts of different freezing times, freezing media, and freezing temperatures were investigated with electron microscopy as well as inductively coupled plasma optical emission spectroscopy. The use of lower freezing temperatures, freezing media with higher heat conductivities, and longer freezing periods led to extremely different network structures with enhanced stability. In detail, materials were created in the shape of lamellar, cellular, and dendritic networks. So far, without changing the building blocks, it was not possible to create the selective morphologies of resulting aerogels in cryoaerogelation. Now, these additive-free approaches enable targeted structuring and will open up new opportunities in the future cryoaerogel design.
Collapse
Affiliation(s)
- Dennis Müller
- Institute
of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Callinstraße 3A, Hannover 30167, Germany
- Laboratory
for Nano and Quantum Engineering, Leibniz
Universität Hannover, Schneiderberg 39, Hannover 30167, Germany
| | - Lars F. Klepzig
- Institute
of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Callinstraße 3A, Hannover 30167, Germany
- Cluster
of Excellence PhoenixD (Photonics, Optics and Engineering −
Innovation Across Disciplines), Hannover 30167, Germany
| | - Anja Schlosser
- Institute
of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Callinstraße 3A, Hannover 30167, Germany
- Laboratory
for Nano and Quantum Engineering, Leibniz
Universität Hannover, Schneiderberg 39, Hannover 30167, Germany
| | - Dirk Dorfs
- Institute
of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Callinstraße 3A, Hannover 30167, Germany
- Laboratory
for Nano and Quantum Engineering, Leibniz
Universität Hannover, Schneiderberg 39, Hannover 30167, Germany
- Cluster
of Excellence PhoenixD (Photonics, Optics and Engineering −
Innovation Across Disciplines), Hannover 30167, Germany
| | - Nadja C. Bigall
- Institute
of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Callinstraße 3A, Hannover 30167, Germany
- Laboratory
for Nano and Quantum Engineering, Leibniz
Universität Hannover, Schneiderberg 39, Hannover 30167, Germany
- Cluster
of Excellence PhoenixD (Photonics, Optics and Engineering −
Innovation Across Disciplines), Hannover 30167, Germany
| |
Collapse
|
31
|
Radhouani H, Correia S, Gonçalves C, Reis RL, Oliveira JM. Synthesis and Characterization of Biocompatible Methacrylated Kefiran Hydrogels: Towards Tissue Engineering Applications. Polymers (Basel) 2021; 13:1342. [PMID: 33923932 PMCID: PMC8072540 DOI: 10.3390/polym13081342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/15/2021] [Accepted: 04/18/2021] [Indexed: 02/07/2023] Open
Abstract
Hydrogel application feasibility is still limited mainly due to their low mechanical strength and fragile nature. Therefore, several physical and chemical cross-linking modifications are being used to improve their properties. In this research, methacrylated Kefiran was synthesized by reacting Kefiran with methacrylic anhydride (MA). The developed MA-Kefiran was physicochemically characterized, and its biological properties evaluated by different techniques. Chemical modification of MA-Kefiran was confirmed by 1H-NMR and FTIR and GPC-SEC showed an average Mw of 793 kDa (PDI 1.3). The mechanical data obtained revealed MA-Kefiran to be a pseudoplastic fluid with an extrusion force of 11.21 ± 2.87 N. Moreover, MA-Kefiran 3D cryogels were successfully developed and fully characterized. Through micro-CT and SEM, the scaffolds revealed high porosity (85.53 ± 0.15%) and pore size (33.67 ± 3.13 μm), thick pore walls (11.92 ± 0.44 μm) and a homogeneous structure. Finally, MA-Kefiran revealed to be biocompatible by presenting no hemolytic activity and an improved cellular function of L929 cells observed through the AlamarBlue® assay. By incorporating methacrylate groups in the Kefiran polysaccharide chain, a MA-Kefiran product was developed with remarkable physical, mechanical, and biological properties, resulting in a promising hydrogel to be used in tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Hajer Radhouani
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; (S.C.); (C.G.); (R.L.R.); (J.M.O.)
- ICVS/3B’s–PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| | - Susana Correia
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; (S.C.); (C.G.); (R.L.R.); (J.M.O.)
- ICVS/3B’s–PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| | - Cristiana Gonçalves
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; (S.C.); (C.G.); (R.L.R.); (J.M.O.)
- ICVS/3B’s–PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; (S.C.); (C.G.); (R.L.R.); (J.M.O.)
- ICVS/3B’s–PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| | - Joaquim M. Oliveira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal; (S.C.); (C.G.); (R.L.R.); (J.M.O.)
- ICVS/3B’s–PT Government Associate Laboratory, Braga, 4805-017 Guimarães, Portugal
| |
Collapse
|
32
|
Abstract
Hydrogels are polymeric networks highly swollen with water. Because of their versatility and properties mimicking biological tissues, they are very interesting for biomedical applications. In this aim, the control of porosity is of crucial importance since it governs the transport properties and influences the fate of cells cultured onto or into the hydrogels. Among the techniques allowing for the elaboration of hydrogels, photopolymerization or photo-cross-linking are probably the most powerful and versatile synthetic routes. This Review aims at giving an overview of the literature dealing with photopolymerized hydrogels for which the generation or characterization of porosity is studied. First, the materials (polymers and photoinitiating systems) used for synthesizing hydrogels are presented. The different ways for generating porosity in the photopolymerized hydrogels are explained, and the characterization techniques allowing adequate study of the porosity are presented. Finally, some applications in the field of controlled release and tissue engineering are reviewed.
Collapse
Affiliation(s)
- Erwan Nicol
- Institut des Molécules et Matériaux du Mans (IMMM), UMR 6283 CNRS Le Mans Université, Avenue Olivier Messiaen, 72085 Cedex 9 Le Mans, France
| |
Collapse
|
33
|
He T, Li B, Colombani T, Joshi-Navare K, Mehta S, Kisiday J, Bencherif SA, Bajpayee AG. Hyaluronic Acid-Based Shape-Memory Cryogel Scaffolds for Focal Cartilage Defect Repair. Tissue Eng Part A 2021; 27:748-760. [PMID: 33108972 DOI: 10.1089/ten.tea.2020.0264] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Traumatic joint injuries can result in significant cartilage defects, which can greatly increase the risk of osteoarthritis development. Due to the limited self-healing capacity of avascular cartilage, tissue engineering approaches are required for filling defects and promoting cartilage regeneration. Current approaches utilize invasive surgical procedures for extraction and implantation of autologous chondrocytes; therefore, injectable biomaterials have gained interest to minimize the risk of infection as well as patient pain and discomfort. In this study, we engineered biomimetic, hyaluronic acid (HA)-based cryogel scaffolds that possess shape-memory properties as they contract and regain their shape after syringe injection to noninvasively fill cartilage defects. The cryogels, fabricated with HA and glycidyl methacrylate at -20°C, resulted in an elastic, macroporous, and highly interconnected network that provided a conducive microenvironment for chondrocytes to remain viable and metabolically active after injection through a syringe needle. Chondrocytes seeded within cryogels and cultured for 15 days exhibited enhanced cell proliferation, metabolism, and production of cartilage extracellular matrix glycosaminoglycans compared with HA-based hydrogels. Furthermore, immunohistochemical staining revealed production of collagen type II from chondrocyte-seeded cryogels, indicating the maintenance of cell phenotype. These results demonstrate the potential of chondrocyte-seeded, HA-based, injectable cryogel scaffolds to promote regeneration of cartilage tissue for nonsurgically invasive defect repair. Impact statement Hyaluronic acid-based shape-memory cryogels provide a conducive microenvironment for chondrocyte adhesion, proliferation, and matrix biosynthesis for use in repair of cartilage defects. Due to their sponge-like elastic properties, cryogels can fully recover their original shape back after injection while not impacting metabolism or viability of encapsulated cells. Clinically, they provide an opportunity for filling focal cartilage defects by using a single, minimally invasive injection of a cell encapsulating biocompatible three-dimensional scaffold that can return to its original structure to fit the defect geometry and enable matrix regeneration.
Collapse
Affiliation(s)
- Tengfei He
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA
| | - Boting Li
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA
| | - Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Kasturi Joshi-Navare
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Shikhar Mehta
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA
| | - John Kisiday
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sidi A Bencherif
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA.,Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Ambika G Bajpayee
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA.,Department of Mechanical Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Upadhyay U, Sreedhar I, Singh SA, Patel CM, Anitha K. Recent advances in heavy metal removal by chitosan based adsorbents. Carbohydr Polym 2021; 251:117000. [DOI: 10.1016/j.carbpol.2020.117000] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 08/15/2020] [Accepted: 08/23/2020] [Indexed: 12/11/2022]
|
35
|
Hydrogel Properties and Their Impact on Regenerative Medicine and Tissue Engineering. Molecules 2020; 25:molecules25245795. [PMID: 33302592 PMCID: PMC7764781 DOI: 10.3390/molecules25245795] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/21/2022] Open
Abstract
Hydrogels (HGs), as three-dimensional structures, are widely used in modern medicine, including regenerative medicine. The use of HGs in wound treatment and tissue engineering is a rapidly developing sector of medicine. The unique properties of HGs allow researchers to easily modify them to maximize their potential. Herein, we describe the physicochemical properties of HGs, which determine their subsequent applications in regenerative medicine and tissue engineering. Examples of chemical modifications of HGs and their applications are described based on the latest scientific reports.
Collapse
|
36
|
Perez-Moreno A, Reyes-Peces MDLV, de los Santos DM, Pinaglia-Tobaruela G, de la Orden E, Vilches-Pérez JI, Salido M, Piñero M, de la Rosa-Fox N. Hydroxyl Groups Induce Bioactivity in Silica/Chitosan Aerogels Designed for Bone Tissue Engineering. In Vitro Model for the Assessment of Osteoblasts Behavior. Polymers (Basel) 2020; 12:E2802. [PMID: 33256226 PMCID: PMC7760707 DOI: 10.3390/polym12122802] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
Silica (SiO2)/chitosan (CS) composite aerogels are bioactive when they are submerged in simulated body fluid (SBF), causing the formation of bone-like hydroxyapatite (HAp) layer. Silica-based hybrid aerogels improve the elastic behavior, and the combined CS modifies the network entanglement as a crosslinking biopolymer. Tetraethoxysilane (TEOS)/CS is used as network precursors by employing a sol-gel method assisted with high power ultrasound (600 W). Upon gelation and aging, gels are dried in supercritical CO2 to obtain monoliths. Thermograms provide information about the condensation of the remaining hydroxyl groups (400-700 °C). This step permits the evaluation of the hydroxyl group's content of 2 to 5 OH nm-2. The formed Si-OH groups act as the inductor of apatite crystal nucleation in SBF. The N2 physisorption isotherms show a hysteresis loop of type H3, characteristic to good interconnected porosity, which facilitates both the bioactivity and the adhesion of osteoblasts cells. After two weeks of immersion in SBF, a layer of HAp microcrystals develops on the surface with a stoichiometric Ca/P molar ratio of 1.67 with spherulite morphology and uniform sizes of 6 μm. This fact asserts the bioactive behavior of these hybrid aerogels. Osteoblasts are cultured on the selected samples and immunolabeled for cytoskeletal and focal adhesion expression related to scaffold nanostructure and composition. The initial osteoconductive response observes points to a great potential of tissue engineering for the designed composite aerogels.
Collapse
Affiliation(s)
- Antonio Perez-Moreno
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (M.d.l.V.R.-P.); (E.d.l.O.); (J.I.V.-P.); (M.S.); (M.P.); (N.d.l.R.-F.)
- Instituto de Microscopía Electrónica y Materiales (IMEYMAT), University of Cadiz, 11510 Cádiz, Spain
- Department of Condensed Matter Physics, Faculty of Science, University of Cadiz, 11510 Cádiz, Spain
| | - María de las Virtudes Reyes-Peces
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (M.d.l.V.R.-P.); (E.d.l.O.); (J.I.V.-P.); (M.S.); (M.P.); (N.d.l.R.-F.)
- Instituto de Microscopía Electrónica y Materiales (IMEYMAT), University of Cadiz, 11510 Cádiz, Spain
- Department of Condensed Matter Physics, Faculty of Science, University of Cadiz, 11510 Cádiz, Spain
| | | | | | - Emilio de la Orden
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (M.d.l.V.R.-P.); (E.d.l.O.); (J.I.V.-P.); (M.S.); (M.P.); (N.d.l.R.-F.)
- Department of Histology, SCIBM, Faculty of Medicine, University of Cadiz, 11004 Cádiz, Spain;
| | - José Ignacio Vilches-Pérez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (M.d.l.V.R.-P.); (E.d.l.O.); (J.I.V.-P.); (M.S.); (M.P.); (N.d.l.R.-F.)
- Department of Histology, SCIBM, Faculty of Medicine, University of Cadiz, 11004 Cádiz, Spain;
| | - Mercedes Salido
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (M.d.l.V.R.-P.); (E.d.l.O.); (J.I.V.-P.); (M.S.); (M.P.); (N.d.l.R.-F.)
- Department of Histology, SCIBM, Faculty of Medicine, University of Cadiz, 11004 Cádiz, Spain;
| | - Manuel Piñero
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (M.d.l.V.R.-P.); (E.d.l.O.); (J.I.V.-P.); (M.S.); (M.P.); (N.d.l.R.-F.)
- Instituto de Microscopía Electrónica y Materiales (IMEYMAT), University of Cadiz, 11510 Cádiz, Spain
- Department of Condensed Matter Physics, Faculty of Science, University of Cadiz, 11510 Cádiz, Spain
| | - Nicolás de la Rosa-Fox
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009 Cádiz, Spain; (M.d.l.V.R.-P.); (E.d.l.O.); (J.I.V.-P.); (M.S.); (M.P.); (N.d.l.R.-F.)
- Instituto de Microscopía Electrónica y Materiales (IMEYMAT), University of Cadiz, 11510 Cádiz, Spain
- Department of Condensed Matter Physics, Faculty of Science, University of Cadiz, 11510 Cádiz, Spain
| |
Collapse
|
37
|
Joshi Navare K, Colombani T, Rezaeeyazdi M, Bassous N, Rana D, Webster T, Memic A, Bencherif SA. Needle-injectable microcomposite cryogel scaffolds with antimicrobial properties. Sci Rep 2020; 10:18370. [PMID: 33110210 PMCID: PMC7591905 DOI: 10.1038/s41598-020-75196-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 10/01/2020] [Indexed: 12/24/2022] Open
Abstract
Porous three-dimensional hydrogel scaffolds have an exquisite ability to promote tissue repair. However, because of their high water content and invasive nature during surgical implantation, hydrogels are at an increased risk of bacterial infection. Recently, we have developed elastic biomimetic cryogels, an advanced type of polymeric hydrogel, that are syringe-deliverable through hypodermic needles. These needle-injectable cryogels have unique properties, including large and interconnected pores, mechanical robustness, and shape-memory. Like hydrogels, cryogels are also susceptible to colonization by microbial pathogens. To that end, our minimally invasive cryogels have been engineered to address this challenge. Specifically, we hybridized the cryogels with calcium peroxide microparticles to controllably produce bactericidal hydrogen peroxide. Our novel microcomposite cryogels exhibit antimicrobial properties and inhibit antibiotic-resistant bacteria (MRSA and Pseudomonas aeruginosa), the most common cause of biomaterial implant failure in modern medicine. Moreover, the cryogels showed negligible cytotoxicity toward murine fibroblasts and prevented activation of primary bone marrow-derived dendritic cells ex vivo. Finally, in vivo data suggested tissue integration, biodegradation, and minimal host inflammatory responses when the antimicrobial cryogels, even when purposely contaminated with bacteria, were subcutaneously injected in mice. Collectively, these needle-injectable microcomposite cryogels show great promise for biomedical applications, especially in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Kasturi Joshi Navare
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | | | - Nicole Bassous
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Devyesh Rana
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Thomas Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
- Wenzhou Institute for Biomaterials and Engineering, Wenzhou, 325001, China
| | - Adnan Memic
- Center of Nanotechnology, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Sidi A Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA.
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
- Sorbonne University, UTC CNRS UMR 7338, Biomechanics and Bioengineering (BMBI), University of Technology of Compiègne, 60203, Compiègne, France.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
38
|
Abstract
A major function of the immune system is to detect threat from foreign invaders, tissue damage, or cancer and to mount a counter response that resolves the threat, restores homeostasis, and supplies immunological memory to prevent a second assault. Our increasing understanding of the immune system has opened up numerous avenues for modulating immune responses against infections, cancer, and autoimmunity. However, agents used for immunomodulation have been traditionally administered systemically via bolus injection, leading to unintended consequences by disrupting homeostasis at nontarget sites. Consequently, systemic hyperactivation and hypoactivation can result from bolus administration of immune-activators and immunosuppressants, respectively. Macroscale biomaterial scaffolds can instead be placed at the intended target site to provide both localized, controlled release of immunomodulatory agents and control over local immune cell trafficking and function, potentially maximizing therapeutic efficacy and limiting systemic exposure. These scaffolds have found utility in the area of cancer immunotherapy, especially in situ cancer vaccination where controlled release of factors such as granulocyte-macrophage colony-stimulating factor (GM-CSF) and the local presentation of tumor antigen and danger signals lead to the recruitment of immature dendritic cells and facilitate their activation and antigen presentation. These cells eventually migrate into secondary lymphoid organs where they prime tumor specific T cells for downstream tumor clearance. Scaffolds can also be used in adoptive T cell therapy to generate large numbers of potent antigen specific T cells or chimeric antigen receptor (CAR) T cells in vitro for subsequent delivery to patients. Macroscale biomaterial scaffolds have also found utility beyond cancer immunotherapy and have been developed to promote immune tolerance by regulatory T cell induction and to expedite tissue regeneration. The design of these macroscale biomaterial scaffolds considers their biocompatibility, biodegradability, mode of delivery, porosity, and kinetics of therapeutic cargo release. Consequently, the numerous approaches that have been developed to fabricate biomaterial scaffolds are aimed at tuning these parameters to achieve the desired therapeutic outcome. This Account will discuss the use of biomaterial scaffolds as niches for immunomodulation and will focus on (1) approaches that have been used to fabricate various biomaterial systems being employed as niches for immunomodulation and (2) how these biomaterial systems have been used to modulate immune responses, specifically in area of cancer immunotherapy, where we will discuss the role of macroscale biomaterial scaffolds for in situ vaccination and in vitro T cell expansion. We will also briefly discuss the utility of biomaterial scaffolds beyond cancer, drawing examples from tolerance and tissue regeneration.
Collapse
Affiliation(s)
- Kwasi Adu-Berchie
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - David J. Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| |
Collapse
|
39
|
Rogers ZJ, Zeevi MP, Koppes R, Bencherif SA. Electroconductive Hydrogels for Tissue Engineering: Current Status and Future Perspectives. Bioelectricity 2020; 2:279-292. [PMID: 34476358 PMCID: PMC8370338 DOI: 10.1089/bioe.2020.0025] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Over the past decade, electroconductive hydrogels, integrating both the biomimetic attributes of hydrogels and the electrochemical properties of conductive materials, have gained significant attention. Hydrogels, three-dimensional and swollen hydrophilic polymer networks, are an important class of tissue engineering (TE) scaffolds owing to their microstructural and mechanical properties, ability to mimic the native extracellular matrix, and promote tissue repair. However, hydrogels are intrinsically insulating and therefore unable to emulate the complex electrophysiological microenvironment of cardiac and neural tissues. To overcome this challenge, electroconductive materials, including carbon-based materials, nanoparticles, and polymers, have been incorporated within nonconductive hydrogels to replicate the electrical and biological characteristics of biological tissues. This review gives a brief introduction on the rational design of electroconductive hydrogels and their current applications in TE, especially for neural and cardiac regeneration. The recent progress and development trends of electroconductive hydrogels, their challenges, and clinical translatability, as well as their future perspectives, with a focus on advanced manufacturing technologies, are also discussed.
Collapse
Affiliation(s)
- Zachary J. Rogers
- Department of Chemical Engineering and Northeastern University, Boston, Massachusetts, USA
| | - Michael P. Zeevi
- Department of Chemical Engineering and Northeastern University, Boston, Massachusetts, USA
| | - Ryan Koppes
- Department of Chemical Engineering and Northeastern University, Boston, Massachusetts, USA
| | - Sidi A. Bencherif
- Department of Chemical Engineering and Northeastern University, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- Biomechanics and Bioengineering (BMBI), UTC CNRS UMR 7338, University of Technology of Compiègne, Compiègne, France
| |
Collapse
|
40
|
Advances in biomaterials for adipose tissue reconstruction in plastic surgery. NANOTECHNOLOGY REVIEWS 2020. [DOI: 10.1515/ntrev-2020-0028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
Adipose tissue reconstruction is an important technique for soft tissue defects caused by facial plastic surgery and trauma. Adipose tissue reconstruction can be repaired by fat transplantation and biomaterial filling, but there are some problems in fat transplantation, such as second operation and limited resources. The application of advanced artificial biomaterials is a promising strategy. In this paper, injectable biomaterials and three-dimensional (3D) tissue-engineered scaffold materials for adipose tissue reconstruction in plastic surgery are reviewed. Injectable biomaterials include natural biomaterials and artificial biomaterials, which generally have problems such as high absorptivity of fillers, repeated injection, and rejection. In recent years, the technology of new 3D tissue-engineering scaffold materials with adipose-derived stem cells (ADSCs) and porous scaffold as the core has made good progress in fat reconstruction, which is expected to solve the current problem of clinical adipose tissue reconstruction, and various biomaterials preparation technology and transformation research also provide the basis for clinical transformation of fat tissue reconstruction.
Collapse
|
41
|
Tang Y, Tong X, Conrad B, Yang F. Injectable and in situ crosslinkable gelatin microribbon hydrogels for stem cell delivery and bone regeneration in vivo. Theranostics 2020; 10:6035-6047. [PMID: 32483436 PMCID: PMC7254995 DOI: 10.7150/thno.41096] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
Rationale: Injectable matrices are highly desirable for stem cell delivery. Previous research has highlighted the benefit of scaffold macroporosity in enhancing stem cell survival and bone regeneration in vivo. However, there remains a lack of injectable and in situ crosslinkable macroporous matrices for stem cell delivery to achieve fast bone regeneration in immunocompetent animal model. The goal of this study is to develop an injectable gelatin-based μRB hydrogel supporting direct cell encapsulation that is available in clinics as macroporous matrices to enhance adipose-derived stromal cell (ASC) survival, engraftment and accelerate bone formation in craniofacial defect mouse. Methods: Injectable and in situ crosslinkable gelatin microribbon (μRB)-based macroporous hydrogels were developed by wet-spinning. Injectability was optimized by varying concentration of glutaraldehyde for intracrosslinking of μRB shape, and fibrinogen coating. The efficacy of injectable μRBs to support ASCs delivery and bone regeneration were further assessed in vivo using an immunocompetent mouse cranial defect model. ASCs survival was evaluated by bioluminescent imaging and bone regeneration was assessed by micro-CT. The degradation and biocompatibility were determined by histological analysis. Results: We first optimized injectability by varying concentration of glutaraldehyde used to fix gelatin μRBs. The injectable μRB formulation were subsequently coated with fibrinogen, which allows in situ crosslinking by thrombin. Fluorescence imaging and histology showed majority of μRBs degraded by the end of 3 weeks. Injectable μRBs supported comparable level of ASC proliferation and bone regeneration as implantable prefabricated μRB controls. Adding low dosage of BMP2 (100 ng per scaffold) with ASCs substantially accelerated the speed of mineralized bone regeneration, with 90% of the bone defect refilled by week 8. Immunostaining showed M1 (pro-inflammatory) macrophages were recruited to the defect at day 3, and was replaced by M2 (anti-inflammatory) macrophages by week 2. Adding μRBs or BMP2 did not alter macrophage response. Injectable µRBs supported vascularization, and BMP-2 further enhanced vascularization. Conclusions: Our results demonstrated that µRB-based scaffolds enhanced ASC survival and accelerated bone regeneration after injection into critical sized cranial defect mouse. Such injectable µRB-based scaffold can provide a versatile biomaterial for delivering various stem cell types and enhancing tissue regeneration.
Collapse
|
42
|
Owen R, Sherborne C, Evans R, Reilly GC, Claeyssens F. Combined Porogen Leaching and Emulsion Templating to produce Bone Tissue Engineering Scaffolds. Int J Bioprint 2020; 6:265. [PMID: 32782992 PMCID: PMC7415854 DOI: 10.18063/ijb.v6i2.265] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/01/2020] [Indexed: 01/13/2023] Open
Abstract
Bone has a hierarchy of porosity that is often overlooked when creating tissue engineering scaffolds where pore sizes are typically confined to a single order of magnitude. High internal phase emulsion (HIPE) templating produces polymerized HIPEs (polyHIPEs): highly interconnected porous polymers which have two length scales of porosity covering the 1-100 μm range. However, additional larger scales of porosity cannot be introduced in the standard emulsion formulation. Researchers have previously overcome this by additively manufacturing emulsions; fabricating highly microporous struts into complex macroporous geometries. This is time consuming and expensive; therefore, here we assessed the feasibility of combining porogen leaching with emulsion templating to introduce additional macroporosity. Alginate beads between 275 and 780 μm were incorporated into the emulsion at 0, 50, and 100 wt%. Once polymerized, alginate was dissolved leaving highly porous polyHIPE scaffolds with added macroporosity. The compressive modulus of the scaffolds decreased as alginate porogen content increased. Cellular performance was assessed using MLO-A5 post-osteoblasts. Seeding efficiency was significantly higher and mineralized matrix deposition was more uniformly deposited throughout porogen leached scaffolds compared to plain polyHIPEs. Deep cell infiltration only occurred in porogen leached scaffolds as detected by histology and lightsheet microscopy. This study reveals a quick, low cost and simple method of producing multiscale porosity scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Robert Owen
- Department of Materials Science and Engineering, INSIGNEO Institute for in silico Medicine, University of Sheffield, UK
- Department of Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, UK
- Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham Biodiscovery Institute, University Park, UK
| | - Colin Sherborne
- Department of Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, UK
| | - Richard Evans
- Bioengineering, Interdisciplinary Programmes Engineering, University of Sheffield, UK
| | - Gwendolen C. Reilly
- Department of Materials Science and Engineering, INSIGNEO Institute for in silico Medicine, University of Sheffield, UK
- Department of Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, UK
| | - Frederik Claeyssens
- Department of Materials Science and Engineering, INSIGNEO Institute for in silico Medicine, University of Sheffield, UK
- Department of Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, UK
| |
Collapse
|
43
|
Haider A, Haider S, Rao Kummara M, Kamal T, Alghyamah AAA, Jan Iftikhar F, Bano B, Khan N, Amjid Afridi M, Soo Han S, Alrahlah A, Khan R. Advances in the scaffolds fabrication techniques using biocompatible polymers and their biomedical application: A technical and statistical review. JOURNAL OF SAUDI CHEMICAL SOCIETY 2020. [DOI: 10.1016/j.jscs.2020.01.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
44
|
Aerts E, Li J, Van Steenbergen MJ, Degrande T, Jansen JA, Walboomers XF. Porous titanium fiber mesh with tailored elasticity and its effect on stromal cells. J Biomed Mater Res B Appl Biomater 2020; 108:2180-2191. [PMID: 31943758 PMCID: PMC7217192 DOI: 10.1002/jbm.b.34556] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 12/05/2019] [Accepted: 01/06/2020] [Indexed: 12/18/2022]
Abstract
Porous titanium fiber mesh (TFM) is considered a suitable scaffold material for bone reconstruction. Also, TFM can be used to cover the surface of bone‐anchored devices, that is, orthopedic or dental implants. The titanium fiber size has an effect of the stiffness as well as porosity of the titanium mesh, which can influence the behavior of bone forming cells. Therefore, the aim of this study was to vary TFM composition, in order to achieve different stiffness, and to assess the effects of such variation on the behavior of bone marrow‐derived stromal cells (BMSCs). With that purpose, nine types of TFM (porosities 60–87%; fiber size 22–50 μm), were examined for their mechanical properties as well as their effect on the proliferation and differentiation of rat bone marrow‐derived stromal cells (rBMSCs) up to 21 days. Dynamic mechanical analysis revealed that the stiffness of TFM were lower than of solid titanium and decreased with larger fiber sizes. The stiffness could effectively be tailored by altering fiber properties, which altered the pore simultaneously. For the 22 and 35 μm size fiber meshes with the highest porosity, the stiffness closely matched the value found in literature for cortical bone. Finally, all tested TFM types supported the growth and differentiation of rBMSCs. We concluded that TFM material has been proven cytocompatible. Further preclinical studies are needed to assess which TFM type is most suitable as clinical use for bone ingrowth and bone regeneration.
Collapse
Affiliation(s)
- Evy Aerts
- Biomaterials, Department of Dentistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jinmeng Li
- Biomaterials, Department of Dentistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mies J Van Steenbergen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | - John A Jansen
- Biomaterials, Department of Dentistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - X Frank Walboomers
- Biomaterials, Department of Dentistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
45
|
Nikolova MP, Chavali MS. Recent advances in biomaterials for 3D scaffolds: A review. Bioact Mater 2019; 4:271-292. [PMID: 31709311 PMCID: PMC6829098 DOI: 10.1016/j.bioactmat.2019.10.005] [Citation(s) in RCA: 427] [Impact Index Per Article: 85.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/07/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023] Open
Abstract
Considering the advantages and disadvantages of biomaterials used for the production of 3D scaffolds for tissue engineering, new strategies for designing advanced functional biomimetic structures have been reviewed. We offer a comprehensive summary of recent trends in development of single- (metal, ceramics and polymers), composite-type and cell-laden scaffolds that in addition to mechanical support, promote simultaneous tissue growth, and deliver different molecules (growth factors, cytokines, bioactive ions, genes, drugs, antibiotics, etc.) or cells with therapeutic or facilitating regeneration effect. The paper briefly focuses on divers 3D bioprinting constructs and the challenges they face. Based on their application in hard and soft tissue engineering, in vitro and in vivo effects triggered by the structural and biological functionalized biomaterials are underlined. The authors discuss the future outlook for the development of bioactive scaffolds that could pave the way for their successful imposing in clinical therapy.
Collapse
Affiliation(s)
- Maria P. Nikolova
- Department of Material Science and Technology, University of Ruse “A. Kanchev”, 8 Studentska Str., 7000, Ruse, Bulgaria
| | - Murthy S. Chavali
- Shree Velagapudi Ramakrishna Memorial College (PG Studies, Autonomous), Nagaram, 522268, Guntur District, India
- PG Department of Chemistry, Dharma Appa Rao College, Nuzvid, 521201, Krishna District, India
- MCETRC, Tenali, 522201, Guntur District, Andhra Pradesh, India
| |
Collapse
|
46
|
Mantha S, Pillai S, Khayambashi P, Upadhyay A, Zhang Y, Tao O, Pham HM, Tran SD. Smart Hydrogels in Tissue Engineering and Regenerative Medicine. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E3323. [PMID: 31614735 PMCID: PMC6829293 DOI: 10.3390/ma12203323] [Citation(s) in RCA: 353] [Impact Index Per Article: 70.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 01/01/2023]
Abstract
The field of regenerative medicine has tremendous potential for improved treatment outcomes and has been stimulated by advances made in bioengineering over the last few decades. The strategies of engineering tissues and assembling functional constructs that are capable of restoring, retaining, and revitalizing lost tissues and organs have impacted the whole spectrum of medicine and health care. Techniques to combine biomimetic materials, cells, and bioactive molecules play a decisive role in promoting the regeneration of damaged tissues or as therapeutic systems. Hydrogels have been used as one of the most common tissue engineering scaffolds over the past two decades due to their ability to maintain a distinct 3D structure, to provide mechanical support for the cells in the engineered tissues, and to simulate the native extracellular matrix. The high water content of hydrogels can provide an ideal environment for cell survival, and structure which mimics the native tissues. Hydrogel systems have been serving as a supportive matrix for cell immobilization and growth factor delivery. This review outlines a brief description of the properties, structure, synthesis and fabrication methods, applications, and future perspectives of smart hydrogels in tissue engineering.
Collapse
Affiliation(s)
- Somasundar Mantha
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Sangeeth Pillai
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Parisa Khayambashi
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Akshaya Upadhyay
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Yuli Zhang
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Owen Tao
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Hieu M Pham
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| |
Collapse
|
47
|
Mandakhbayar N, El-Fiqi A, Lee JH, Kim HW. Evaluation of Strontium-Doped Nanobioactive Glass Cement for Dentin–Pulp Complex Regeneration Therapy. ACS Biomater Sci Eng 2019; 5:6117-6126. [DOI: 10.1021/acsbiomaterials.9b01018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Nandin Mandakhbayar
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, South Korea
| | - Ahmed El-Fiqi
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, South Korea
- Glass Research Department, National Research Centre, Cairo 12622, Egypt
| | - Jung-Hwan Lee
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, South Korea
- Glass Research Department, National Research Centre, Cairo 12622, Egypt
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 330-714, South Korea
| | - Hae-Won Kim
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, South Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 330-714, South Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
48
|
Huynh C, Shih TY, Mammoo A, Samant A, Pathan S, Nelson DW, Ferran C, Mooney D, LoGerfo F, Pradhan-Nabzdyk L. Delivery of targeted gene therapies using a hybrid cryogel-coated prosthetic vascular graft. PeerJ 2019; 7:e7377. [PMID: 31497383 PMCID: PMC6707340 DOI: 10.7717/peerj.7377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/28/2019] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES The success of prosthetic vascular grafts in the management of peripheral arterial disease is frequently limited by the development of anastomotic neointimal hyperplasia (ANIH), with the host response to prosthetic grafts beginning soon after implantation. To address this, we combine a platform of polyethylene terephthalate (PET) fabric with an applied cryogel layer containing biologic agents to create a bioactive prosthetic graft system, with the ability to deliver therapeutics targeting modulators of the ANIH-associated transcriptome response, along with antithrombotic agents. METHODS Hybrid graft materials were synthesized by cryopolymerization of methacrylated alginate and heparin onto electrospun (ePET), knitted PET (kPET), or woven PET (wPET). Arg-Gly-Asp (RGD) peptides were added to increase cell adhesion. Scanning electron microscopy (SEM) was used to study the microstructure at 1 day, and 2, 4, and 8 weeks. Physical properties such as swelling ratio, pore connectivity, shape recovery, and stiffness were evaluated. Human aortic endothelial cell (HAoEC) adherence was visualized using confocal microscopy after 24 hours and proliferation was evaluated with a resazurin-based assay for 7 days. Confocal microscopy was used to assess delivery of adeno-associated virus (AAV-GFP) after incubation of hybrid grafts with HAoECs. Heparin activity of the materials was measured using an anti-Xa assay. RESULTS SEM demonstrated large interconnected pores throughout the entire structure for all graft types, with minimal degradation of the cryogel after 8 weeks. Hybrid materials showed a trend towards increased shape recovery, increased stiffness, decreased swelling ratio, and no difference in pore connectivity. HAoECs incorporated, adhered, and proliferated over 7 days on all materials. HAoECs were successfully transduced with AAV-GFP from the hybrid graft materials. Anti-Xa assay confirmed continued activity of heparin from all materials for over 7 days. CONCLUSIONS We have developed a bioactive prosthetic graft system with a cryogel coating capable of delivering biologic agents with antithrombotic activity. By applying the cryogel and selected agents onto PET prior to graft implantation, this study sets the stage for the system to be individualized and tailored to the patient, with bioengineering and targeted gene therapy strategies dovetailing to create an improved prosthetic graft adaptable to emerging knowledge and technologies.
Collapse
Affiliation(s)
- Cindy Huynh
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- Department of Surgery, State University of New York (SUNY), Syracuse, NY, United States of America
| | - Ting-Yu Shih
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Alexander Mammoo
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
- Division of Pharmacology, Department of Pharmaceutical Biosciences, Uppsala Universitet, Uppsala, Sweden
| | - Amruta Samant
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Saif Pathan
- BioSurfaces, Inc, Ashland, MA, United States of America
| | | | - Christiane Ferran
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - David Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Frank LoGerfo
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Leena Pradhan-Nabzdyk
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| |
Collapse
|
49
|
Li Y, Liao C, Tjong SC. Electrospun Polyvinylidene Fluoride-Based Fibrous Scaffolds with Piezoelectric Characteristics for Bone and Neural Tissue Engineering. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E952. [PMID: 31261995 PMCID: PMC6669491 DOI: 10.3390/nano9070952] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/14/2019] [Accepted: 06/15/2019] [Indexed: 02/07/2023]
Abstract
Polyvinylidene fluoride (PVDF) and polyvinylidene fluoride-trifluoroethylene (P(VDF-TrFE) with excellent piezoelectricity and good biocompatibility are attractive materials for making functional scaffolds for bone and neural tissue engineering applications. Electrospun PVDF and P(VDF-TrFE) scaffolds can produce electrical charges during mechanical deformation, which can provide necessary stimulation for repairing bone defects and damaged nerve cells. As such, these fibrous mats promote the adhesion, proliferation and differentiation of bone and neural cells on their surfaces. Furthermore, aligned PVDF and P(VDF-TrFE) fibrous mats can enhance neurite growth along the fiber orientation direction. These beneficial effects derive from the formation of electroactive, polar β-phase having piezoelectric properties. Polar β-phase can be induced in the PVDF fibers as a result of the polymer jet stretching and electrical poling during electrospinning. Moreover, the incorporation of TrFE monomer into PVDF can stabilize the β-phase without mechanical stretching or electrical poling. The main drawbacks of electrospinning process for making piezoelectric PVDF-based scaffolds are their small pore sizes and the use of highly toxic organic solvents. The small pore sizes prevent the infiltration of bone and neuronal cells into the scaffolds, leading to the formation of a single cell layer on the scaffold surfaces. Accordingly, modified electrospinning methods such as melt-electrospinning and near-field electrospinning have been explored by the researchers to tackle this issue. This article reviews recent development strategies, achievements and major challenges of electrospun PVDF and P(VDF-TrFE) scaffolds for tissue engineering applications.
Collapse
Affiliation(s)
- Yuchao Li
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng 252000, China.
| | - Chengzhu Liao
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Sie Chin Tjong
- Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China.
| |
Collapse
|
50
|
Conrad B, Han LH, Yang F. Gelatin-Based Microribbon Hydrogels Accelerate Cartilage Formation by Mesenchymal Stem Cells in Three Dimensions. Tissue Eng Part A 2019; 24:1631-1640. [PMID: 29926770 DOI: 10.1089/ten.tea.2018.0011] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hydrogels (HGs) are attractive matrices for cell-based cartilage tissue regeneration given their injectability and ability to fill defects with irregular shapes. However, most HGs developed to date often lack cell scale macroporosity, which restrains the encapsulated cells, leading to delayed new extracellular matrix deposition restricted to pericellular regions. Furthermore, tissue-engineered cartilage using conventional HGs generally suffers from poor mechanical property and fails to restore the load-bearing property of articular cartilage. The goal of this study was to evaluate the potential of macroporous gelatin-based microribbon (μRB) HGs as novel 3D matrices for accelerating chondrogenesis and new cartilage formation by human mesenchymal stem cells (MSCs) in 3D with improved mechanical properties. Unlike conventional HGs, these μRB HGs are inherently macroporous and exhibit cartilage-mimicking shock-absorbing mechanical property. After 21 days of culture, MSC-seeded μRB scaffolds exhibit a 20-fold increase in compressive modulus to 225 kPa, a range that is approaching the level of native cartilage. In contrast, HGs only resulted in a modest increase in compressive modulus of 65 kPa. Compared with conventional HGs, macroporous μRB scaffolds significantly increased the total amount of neocartilage produced by MSCs in 3D, with improved interconnectivity and mechanical strength. Altogether, these results validate gelatin-based μRBs as promising scaffolds for enhancing and accelerating MSC-based cartilage regeneration and may be used to enhance cartilage regeneration using other cell types as well.
Collapse
Affiliation(s)
- Bogdan Conrad
- 1 Program of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Li-Hsin Han
- 2 Department of Orthopedic Surgery, Stanford University School of Medicine , Stanford, California
| | - Fan Yang
- 3 Department of Orthopedic Surgery and Bioengineering, Stanford University , Stanford, California
| |
Collapse
|