1
|
Yin W, Jiang Y, Ma G, Mbituyimana B, Xu J, Shi Z, Yang G, Chen H. A review: Carrier-based hydrogels containing bioactive molecules and stem cells for ischemic stroke therapy. Bioact Mater 2025; 49:39-62. [PMID: 40124600 PMCID: PMC11928985 DOI: 10.1016/j.bioactmat.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/22/2024] [Accepted: 01/13/2025] [Indexed: 03/25/2025] Open
Abstract
Ischemic stroke (IS), a cerebrovascular disease, is the leading cause of physical disability and death worldwide. Tissue plasminogen activator (tPA) and thrombectomy are limited by a narrow therapeutic time window. Although strategies such as drug therapies and cellular therapies have been used in preclinical trials, some important issues in clinical translation have not been addressed: low stem cell survival and drug delivery limited by the blood-brain barrier (BBB). Among the therapeutic options currently sought, carrier-based hydrogels hold great promise for the repair and regeneration of neural tissue in the treatment of ischemic stroke. The advantage lies in the ability to deliver drugs and cells to designated parts of the brain in an injectable manner to enhance therapeutic efficacy. Here, this article provides an overview of the use of carrier-based hydrogels in ischemic stroke therapy and focuses on the use of hydrogel scaffolds containing bioactive molecules and stem cells. In addition to this, we provide a more in-depth summary of the composition, physicochemical properties and physiological functions of the materials themselves. Finally, we also outline the prospects and challenges for clinical translation of hydrogel therapy for IS.
Collapse
Affiliation(s)
- Wenqi Yin
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuchi Jiang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Guangrui Ma
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bricard Mbituyimana
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jia Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhijun Shi
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
2
|
Xue B, Xu Z, Li L, Guo K, Mi J, Wu H, Li Y, Xie C, Jin J, Xu J, Jiang C, Gu X, Qin M, Jiang Q, Cao Y, Wang W. Hydrogels with programmed spatiotemporal mechanical cues for stem cell-assisted bone regeneration. Nat Commun 2025; 16:3633. [PMID: 40240370 PMCID: PMC12003706 DOI: 10.1038/s41467-025-59016-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
Hydrogels are extensively utilized in stem cell-based tissue regeneration, providing a supportive environment that facilitates cell survival, differentiation, and integration with surrounding tissues. However, designing hydrogels for regenerating hard tissues like bone presents significant challenges. Here, we introduce macroporous hydrogels with spatiotemporally programmed mechanical properties for stem cell-driven bone regeneration. Using liquid-liquid phase separation and interfacial supramolecular self-assembly of protein fibres, the macroporous structure of hydrogels provide ample space to prevent contact inhibition during proliferation. The rigid protein fibre-coated pore shell provides sustained mechanical cues for guiding osteodifferentiation and protecting against mechanical loads. Temporally, the hydrogel exhibits tunable degradation rates that can synchronize with new tissue deposition to some extent. By integrating localized mechanical heterogeneity, macroporous structures, surface chemistry, and regenerative degradability, we demonstrate the efficacy of these stem cell-encapsulated hydrogels in rabbit and porcine models. This marks a substantial advancement in tailoring the mechanical properties of hydrogels for stem cell-assisted tissue regeneration.
Collapse
Affiliation(s)
- Bin Xue
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| | - Zhengyu Xu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), MOE Key Laboratory of High Performance Polymer Materials and Technology, and State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China
| | - Kaiqiang Guo
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Jing Mi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Haipeng Wu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Yiran Li
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Chunmei Xie
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jing Jin
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Juan Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chunping Jiang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Xiaosong Gu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Meng Qin
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, China.
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
- Chemistry and Biomedicine Innovation Center (ChemBIC), MOE Key Laboratory of High Performance Polymer Materials and Technology, and State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| | - Wei Wang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| |
Collapse
|
3
|
Chen K, Liu Z, Zhou X, Zheng W, Cao H, Yang Z, Wang Z, Ning C, Li Q, Zhao H. Hierarchy Reproduction: Multiphasic Strategies for Tendon/Ligament-Bone Junction Repair. Biomater Res 2025; 29:0132. [PMID: 39844867 PMCID: PMC11751208 DOI: 10.34133/bmr.0132] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/15/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Tendon/ligament-bone junctions (T/LBJs) are susceptible to damage during exercise, resulting in anterior cruciate ligament rupture or rotator cuff tear; however, their intricate hierarchical structure hinders self-regeneration. Multiphasic strategies have been explored to fuel heterogeneous tissue regeneration and integration. This review summarizes current multiphasic approaches for rejuvenating functional gradients in T/LBJ healing. Synthetic, natural, and organism-derived materials are available for in vivo validation. Both discrete and gradient layouts serve as sources of inspiration for organizing specific cues, based on the theories of biomaterial topology, biochemistry, mechanobiology, and in situ delivery therapy, which form interconnected network within the design. Novel engineering can be constructed by electrospinning, 3-dimensional printing, bioprinting, textiling, and other techniques. Despite these efforts being limited at present stage, multiphasic scaffolds show great potential for precise reproduction of native T/LBJs and offer promising solutions for clinical dilemmas.
Collapse
Affiliation(s)
- Kaiting Chen
- Academy of Orthopedics, Guangdong Province, Orthopedic Hospital of Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510665, P. R. China
| | - Zezheng Liu
- Academy of Orthopedics, Guangdong Province, Orthopedic Hospital of Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510665, P. R. China
| | - Xinying Zhou
- Academy of Orthopedics, Guangdong Province, Orthopedic Hospital of Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510665, P. R. China
| | - Wanyu Zheng
- School of Pharmaceutical Sciences,
Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - He Cao
- Academy of Orthopedics, Guangdong Province, Orthopedic Hospital of Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510665, P. R. China
| | - Zijian Yang
- Academy of Orthopedics, Guangdong Province, Orthopedic Hospital of Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510665, P. R. China
| | - Zhengao Wang
- School of Materials Science and Engineering,
South China University of Technology, Guangzhou 510006, P. R. China
| | - Chengyun Ning
- School of Materials Science and Engineering,
South China University of Technology, Guangzhou 510006, P. R. China
| | - Qingchu Li
- Academy of Orthopedics, Guangdong Province, Orthopedic Hospital of Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510665, P. R. China
| | - Huiyu Zhao
- Academy of Orthopedics, Guangdong Province, Orthopedic Hospital of Guangdong Province,
The Third Affiliated Hospital of Southern Medical University, Guangzhou 510665, P. R. China
| |
Collapse
|
4
|
Lipari S, Sacco P, Cok M, Scognamiglio F, Romano M, Brun F, Giulianini PG, Marsich E, Aachmann FL, Donati I. Hydrogel Elastic Energy: A Stressor Triggering an Adaptive Stress-Mediated Cell Response. Adv Healthc Mater 2025; 14:e2402400. [PMID: 39535422 PMCID: PMC11730662 DOI: 10.1002/adhm.202402400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The crosstalk between the cells and the extracellular matrix (ECM) is bidirectional and consists of a pushing/pulling stretch exerted by the cells and a mechanical resistance counteracted by the surrounding microenvironment. It is widely recognized that the stiffness of the ECM, its viscoelasticity, and its overall deformation are the most important traits influencing the response of the cells. Here these three parameters are combined into a concept of elastic energy, which in biological terms represents the mechanical feedback that cells perceive when the ECM is deformed. It is shown that elastic energy is a stress factor that influences the response of cells in three-dimensional (3D) cultures. Strikingly, the higher the elastic energy of the matrix and thus the mechanical feedback, the higher the stress state of the cells, which correlates with the formation of G3BP-mediated stress granules. This condition is associated with an increase in alkaline phosphatase (ALP) activity but a decrease in gene expression and is mediated by the nuclear translocation of Yes-associated protein (YAP). This work supports the importance of considering the elastic energy as mechano-controller in regulating cellular stress state in 3D cultures.
Collapse
Affiliation(s)
- Sara Lipari
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Pasquale Sacco
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Michela Cok
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | | | - Maurizio Romano
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Francesco Brun
- Department of Engineering and ArchitectureUniversity of TriesteVia A. Valerio 6/1Trieste34127Italy
| | | | - Eleonora Marsich
- Department of MedicineSurgery and Health SciencesUniversity of TriestePiazza dell'Ospitale 1Trieste34129Italy
| | - Finn L. Aachmann
- Department of Biotechnology and Food ScienceNorwegian Biopolymer Laboratory (NOBIPOL)NTNU Norwegian University of Science and TechnologySem Sælands vei 6/8Trondheim7491Norway
| | - Ivan Donati
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| |
Collapse
|
5
|
Ehlen Q, Costello JP, Mirsky NA, Slavin BV, Parra M, Ptashnik A, Nayak VV, Coelho PG, Witek L. Treatment of Bone Defects and Nonunion via Novel Delivery Mechanisms, Growth Factors, and Stem Cells: A Review. ACS Biomater Sci Eng 2024; 10:7314-7336. [PMID: 39527574 PMCID: PMC11632667 DOI: 10.1021/acsbiomaterials.4c01279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Bone nonunion following a fracture represents a significant global healthcare challenge, with an overall incidence ranging between 2 and 10% of all fractures. The management of nonunion is not only financially prohibitive but often necessitates invasive surgical interventions. This comprehensive manuscript aims to provide an extensive review of the published literature involving growth factors, stem cells, and novel delivery mechanisms for the treatment of fracture nonunion. Key growth factors involved in bone healing have been extensively studied, including bone morphogenic protein (BMP), vascular endothelial growth factor (VEGF), and platelet-derived growth factor. This review includes both preclinical and clinical studies that evaluated the role of growth factors in acute and chronic nonunion. Overall, these studies revealed promising bridging and fracture union rates but also elucidated complications such as heterotopic ossification and inferior mechanical properties associated with chronic nonunion. Stem cells, particularly mesenchymal stem cells (MSCs), are an extensively studied topic in the treatment of nonunion. A literature search identified articles that demonstrated improved healing responses, osteogenic capacity, and vascularization of fractures due to the presence of MSCs. Furthermore, this review addresses novel mechanisms and materials being researched to deliver these growth factors and stem cells to nonunion sites, including natural/synthetic polymers and bioceramics. The specific mechanisms explored in this review include BMP-induced osteoblast differentiation, VEGF-mediated angiogenesis, and the role of MSCs in multilineage differentiation and paracrine signaling. While these therapeutic modalities exhibit substantial preclinical promise in treating fracture nonunion, there remains a need for further research, particularly in chronic nonunion and large animal models. This paper seeks to identify such translational hurdles which must be addressed in order to progress the aforementioned treatments from the lab to the clinical setting.
Collapse
Affiliation(s)
- Quinn
T. Ehlen
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Joseph P. Costello
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Nicholas A. Mirsky
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Blaire V. Slavin
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Marcelo Parra
- Center
of Excellence in Morphological and Surgical Studies (CEMyQ), Faculty
of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
- Department
of Comprehensive Adult Dentistry, Faculty of Dentistry, Universidad de La Frontera, Temuco 4811230, Chile
| | - Albert Ptashnik
- Biomaterials
Division, NYU Dentistry, New York, New York 10010, United States
| | - Vasudev Vivekanand Nayak
- Department
of Biochemistry and Molecular Biology, University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Paulo G. Coelho
- Department
of Biochemistry and Molecular Biology, University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
- Division
of Plastic Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Lukasz Witek
- Biomaterials
Division, NYU Dentistry, New York, New York 10010, United States
- Department
of Biomedical Engineering, NYU Tandon School
of Engineering, Brooklyn, New York 11201, United States
- Hansjörg
Wyss Department of Plastic Surgery, NYU
Grossman School of Medicine, New
York, New York 10016, United States
| |
Collapse
|
6
|
Cojocaru E, Oprea M, Vlăsceanu GM, Nicolae MC, Popescu RC, Mereuţă PE, Toader AG, Ioniţă M. Dual nanofiber and graphene reinforcement of 3D printed biomimetic supports for bone tissue repair. RSC Adv 2024; 14:32517-32532. [PMID: 39411258 PMCID: PMC11474446 DOI: 10.1039/d4ra06167e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
Replicating the intricate architecture of the extracellular matrix (ECM) is an actual challenge in the field of bone tissue engineering. In the present research study, calcium alginate/cellulose nanofibrils-based 3D printed scaffolds, double-reinforced with chitosan/polyethylene oxide electrospun nanofibers (NFs) and graphene oxide (GO) were prepared using the 3D printing technique. The porous matrix was provided by the calcium alginate, while the anisotropy degree and mechanical properties were ensured by the addition of fillers with different sizes and shapes (CNFs, NFs, GO), similar to the components naturally found in bone ECM. Surface morphology and 3D internal microstructure were analyzed using scanning electron microscopy (SEM) and micro-computed tomography (μ-CT), which evidenced a synergistic effect of the reinforcing and functional fibers addition, as well as of the GO sheets that seem to govern materials structuration. Also, the nanoindentation measurements showed significant differences in the elasticity and viscosity modulus, depending on the measurement point, this supported the anisotropic character of the scaffolds. In vitro assays performed on MG-63 osteoblast cells confirmed the biocompatibility of the calcium alginate-based scaffolds and highlighted the osteostimulatory and mineralization enhancement effect of GO. In virtue of their biocompatibility, structural complexity similar with the one of native bone ECM, and biomimetic mechanical characteristics (e.g. high mechanical strength, durotaxis), these novel materials were considered appropriate for specific functional needs, like guided support for bone tissue formation.
Collapse
Affiliation(s)
- Elena Cojocaru
- Advanced Polymer Materials Group, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
| | - Mădălina Oprea
- Advanced Polymer Materials Group, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
- Faculty of Medical Engineering, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
| | - George Mihail Vlăsceanu
- Advanced Polymer Materials Group, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
- Faculty of Medical Engineering, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
| | - Mădălina-Cristina Nicolae
- Advanced Polymer Materials Group, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
| | - Roxana-Cristina Popescu
- Faculty of Medical Engineering, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
- National Institute for Research and Development in Physics and Nuclear Engineering "Horia Hulubei", Department of Life and Environmental Physics 30 Reactor. Street Magurele Romania
| | - Paul-Emil Mereuţă
- National Institute for Research and Development in Physics and Nuclear Engineering "Horia Hulubei", Department of Applied Nuclear Physics 30 Reactor. Street Magurele Romania
| | - Alin-Georgian Toader
- Advanced Polymer Materials Group, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
- Faculty of Medical Engineering, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
| | - Mariana Ioniţă
- Advanced Polymer Materials Group, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
- Faculty of Medical Engineering, National University of Science and Technology POLITEHNICA Bucharest 1-7 Gh. Polizu Street Bucharest 011061 Romania
- Center of Excellence in Bioengineering, National University of Science and Technology POLITEHNICA Bucharest 6 Iuliu Maniu Boulevard, Campus Building Bucharest 061344 Romania
| |
Collapse
|
7
|
Shen S, Lin Y, Sun J, Liu Y, Chen Y, Lu J. A New Tissue Engineering Strategy to Promote Tendon-bone Healing: Regulation of Osteogenic and Chondrogenic Differentiation of Tendon-derived Stem Cells. Orthop Surg 2024; 16:2311-2325. [PMID: 39043618 PMCID: PMC11456719 DOI: 10.1111/os.14152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
In the field of sports medicine, repair surgery for anterior cruciate ligament (ACL) and rotator cuff (RC) injuries are remarkably common. Despite the availability of relatively effective treatment modalities, outcomes often fall short of expectations. This comprehensive review aims to thoroughly examine current strategies employed to promote tendon-bone healing and analyze pertinent preclinical and clinical research. Amidst ongoing investigations, tendon-derived stem cells (TDSCs), which have comparatively limited prior exploration, have garnered increasing attention in the context of tendon-bone healing, emerging as a promising cell type for regenerative therapies. This review article delves into the potential of combining TDSCs with tissue engineering methods, with ACL reconstruction as the main focus. It comprehensively reviews relevant research on ACL and RC healing to address the issues of graft healing and bone tunnel integration. To optimize tendon-bone healing outcomes, our emphasis lies in not only reconstructing the original microstructure of the tendon-bone interface but also achieving proper bone tunnel integration, encompassing both cartilage and bone formation. In this endeavor, we thoroughly analyze the transcriptional and molecular regulatory variables governing TDSCs differentiation, incorporating a retrospective analysis utilizing single-cell sequencing, with the aim of unearthing relevant signaling pathways and processes. By presenting a novel strategy rooted in TDSCs-driven osteogenic and chondrogenic differentiation for tendon-bone healing, this study paves the way for potential future research avenues and promising therapeutic applications. It is anticipated that the findings herein will contribute to advancing the field of tendon-bone healing and foster the exploration of TDSCs as a viable option for regenerative therapies in the future.
Collapse
Affiliation(s)
- Sinuo Shen
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Yucheng Lin
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Jiachen Sun
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Yuanhao Liu
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Yuzhi Chen
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Jun Lu
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| |
Collapse
|
8
|
López-Serrano C, Côté-Paradis Y, Habenstein B, Loquet A, Le Coz C, Ruel J, Laroche G, Durrieu MC. Integrating Mechanics and Bioactivity: A Detailed Assessment of Elasticity and Viscoelasticity at Different Scales in 2D Biofunctionalized PEGDA Hydrogels for Targeted Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:39165-39180. [PMID: 39041490 PMCID: PMC11600396 DOI: 10.1021/acsami.4c10755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Methods for promoting and controlling the differentiation of human mesenchymal stem cells (hMSCs) in vitro before in vivo transplantation are crucial for the advancement of tissue engineering and regenerative medicine. In this study, we developed poly(ethylene glycol) diacrylate (PEGDA) hydrogels with tunable mechanical properties, including elasticity and viscoelasticity, coupled with bioactivity achieved through the immobilization of a mixture of RGD and a mimetic peptide of the BMP-2 protein. Despite the key relevance of hydrogel mechanical properties for cell culture, a standard for its characterization has not been proposed, and comparisons between studies are challenging due to the different techniques employed. Here, a comprehensive approach was employed to characterize the elasticity and viscoelasticity of these hydrogels, integrating compression testing, rheology, and atomic force microscopy (AFM) microindentation. Distinct mechanical behaviors were observed across different PEGDA compositions, and some consistent trends across multiple techniques were identified. Using a photoactivated cross-linker, we controlled the functionalization density independently of the mechanical properties. X-ray photoelectrin spectroscopy and fluorescence microscopy were employed to evaluate the functionalization density of the materials before the culturing of hMSCs on them. The cells cultured on all functionalized hydrogels expressed an early osteoblast marker (Runx2) after 2 weeks, even in the absence of a differentiation-inducing medium compared to our controls. Additionally, after only 1 week of culture with osteogenic differentiation medium, cells showed accelerated differentiation, with clear morphological differences observed among cells in the different conditions. Notably, cells on stiff but stress-relaxing hydrogels exhibited an overexpression of the osteocyte marker E11. This suggests that the combination of the functionalization procedure with the mechanical properties of the hydrogel provides a potent approach to promoting the osteogenic differentiation of hMSCs.
Collapse
Affiliation(s)
- Cristina López-Serrano
- Univ.
Bordeaux, CNRS, Bordeaux INP, CBMN, UMR
5248, Pessac 33600, France
- Laboratoire
d’Ingénierie de Surface, Centre de Recherche sur les
Matériaux Avancés, Département de Génie
des Mines, de la Métallurgie et des Matériaux, Université Laval, Québec, QC G1 V 0A6, Canada
- Axe
médecine régénératrice, Centre de Recherche
du Centre Hospitalier Universitaire de Québec, Hôpital
St-François d’Assise, Québec, QC G1L
3L5, Canada
| | - Yeva Côté-Paradis
- Laboratoire
d’Ingénierie de Surface, Centre de Recherche sur les
Matériaux Avancés, Département de Génie
des Mines, de la Métallurgie et des Matériaux, Université Laval, Québec, QC G1 V 0A6, Canada
- Axe
médecine régénératrice, Centre de Recherche
du Centre Hospitalier Universitaire de Québec, Hôpital
St-François d’Assise, Québec, QC G1L
3L5, Canada
| | - Birgit Habenstein
- Univ.
Bordeaux, CNRS, INSERM, IECB, US1, UAR 3033, F-33600 Pessac, France
| | - Antoine Loquet
- Univ.
Bordeaux, CNRS, INSERM, IECB, US1, UAR 3033, F-33600 Pessac, France
| | - Cédric Le Coz
- Univ.
Bordeaux, CNRS, Bordeaux INP, LCPO, UMR
5629, F-33600 Pessac, France
| | - Jean Ruel
- Département
de Génie Mécanique, Université
Laval, Québec, QC G1V 0A6, Canada
| | - Gaétan Laroche
- Laboratoire
d’Ingénierie de Surface, Centre de Recherche sur les
Matériaux Avancés, Département de Génie
des Mines, de la Métallurgie et des Matériaux, Université Laval, Québec, QC G1 V 0A6, Canada
- Axe
médecine régénératrice, Centre de Recherche
du Centre Hospitalier Universitaire de Québec, Hôpital
St-François d’Assise, Québec, QC G1L
3L5, Canada
| | | |
Collapse
|
9
|
Shaygani H, Shamloo A, Akbarnataj K, Maleki S. In vitro and in vivo investigation of chitosan/silk fibroin injectable interpenetrating network hydrogel with microspheres for cartilage regeneration. Int J Biol Macromol 2024; 270:132126. [PMID: 38723805 DOI: 10.1016/j.ijbiomac.2024.132126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 06/05/2024]
Abstract
Articular cartilage is an avascular and almost acellular tissue with limited self-regenerating capabilities. Although injectable hydrogels have garnered a lot of attention as a promising treatment, a biocompatible hydrogel with adequate mechanical properties is yet to be created. In this study, an interpenetrating network hydrogel comprised of chitosan and silk fibroin was created through electrostatic and hydrophobic bonds, respectively. The polymeric network of the scaffold combined an effective microenvironment for cell activity with enhanced mechanical properties to address the current issues in cartilage scaffolds. Furthermore, microspheres (MS) were utilized for a controlled release of methylprednisolone acetate (MPA), around ~75 % after 35 days. The proposed scaffolds demonstrated great mechanical stability with ~0.047 MPa compressive moduli and ~145 kPa compressive strength. Moreover, the degradation rate of the samples (~45 % after 35 days) was optimized to match neo-cartilage formation. Furthermore, the use of natural biomaterials yielded good biocompatibility with ~76 % chondrocyte viability after 7 days. According to gross observation after 12 weeks the defect site of the treated groups was filled with minimally discernible boundary. These results were confirmed by histopathology assays were the treated groups showed higher chondrocyte count and collagen type II expression.
Collapse
Affiliation(s)
- Hossein Shaygani
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran
| | - Amir Shamloo
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran.
| | - Kazem Akbarnataj
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Department of Mechanical Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Sasan Maleki
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
10
|
Abbadessa A, Ronca A, Salerno A. Integrating bioprinting, cell therapies and drug delivery towards in vivo regeneration of cartilage, bone and osteochondral tissue. Drug Deliv Transl Res 2024; 14:858-894. [PMID: 37882983 DOI: 10.1007/s13346-023-01437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/27/2023]
Abstract
The biological and biomechanical functions of cartilage, bone and osteochondral tissue are naturally orchestrated by a complex crosstalk between zonally dependent cells and extracellular matrix components. In fact, this crosstalk involves biomechanical signals and the release of biochemical cues that direct cell fate and regulate tissue morphogenesis and remodelling in vivo. Three-dimensional bioprinting introduced a paradigm shift in tissue engineering and regenerative medicine, since it allows to mimic native tissue anisotropy introducing compositional and architectural gradients. Moreover, the growing synergy between bioprinting and drug delivery may enable to replicate cell/extracellular matrix reciprocity and dynamics by the careful control of the spatial and temporal patterning of bioactive cues. Although significant advances have been made in this direction, unmet challenges and open research questions persist. These include, among others, the optimization of scaffold zonality and architectural features; the preservation of the bioactivity of loaded active molecules, as well as their spatio-temporal release; the in vitro scaffold maturation prior to implantation; the pros and cons of each animal model and the graft-defect mismatch; and the in vivo non-invasive monitoring of new tissue formation. This work critically reviews these aspects and reveals the state of the art of using three-dimensional bioprinting, and its synergy with drug delivery technologies, to pattern the distribution of cells and/or active molecules in cartilage, bone and osteochondral engineered tissues. Most notably, this work focuses on approaches, technologies and biomaterials that are currently under in vivo investigations, as these give important insights on scaffold performance at the implantation site and its interaction/integration with surrounding tissues.
Collapse
Affiliation(s)
- Anna Abbadessa
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), IDIS Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Campus Vida, Santiago de Compostela, Spain.
| | - Alfredo Ronca
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125, Naples, Italy.
| | - Aurelio Salerno
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, 80125, Naples, Italy.
| |
Collapse
|
11
|
Jalloh US, Gsell A, Gultian KA, MacAulay J, Madden A, Smith J, Siri L, Vega SL. Synthesis and Photopatterning of Synthetic Thiol-Norbornene Hydrogels. Gels 2024; 10:164. [PMID: 38534582 DOI: 10.3390/gels10030164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Hydrogels are a class of soft biomaterials and the material of choice for a myriad of biomedical applications due to their biocompatibility and highly tunable mechanical and biochemical properties. Specifically, light-mediated thiol-norbornene click reactions between norbornene-modified macromers and di-thiolated crosslinkers can be used to form base hydrogels amenable to spatial biochemical modifications via subsequent light reactions between pendant norbornenes in the hydrogel network and thiolated peptides. Macromers derived from natural sources (e.g., hyaluronic acid, gelatin, alginate) can cause off-target cell signaling, and this has motivated the use of synthetic macromers such as poly(ethylene glycol) (PEG). In this study, commercially available 8-arm norbornene-modified PEG (PEG-Nor) macromers were reacted with di-thiolated crosslinkers (dithiothreitol, DTT) to form synthetic hydrogels. By varying the PEG-Nor weight percent or DTT concentration, hydrogels with a stiffness range of 3.3 kPa-31.3 kPa were formed. Pendant norbornene groups in these hydrogels were used for secondary reactions to either increase hydrogel stiffness (by reacting with DTT) or to tether mono-thiolated peptides to the hydrogel network. Peptide functionalization has no effect on bulk hydrogel mechanics, and this confirms that mechanical and biochemical signals can be independently controlled. Using photomasks, thiolated peptides can also be photopatterned onto base hydrogels, and mesenchymal stem cells (MSCs) attach and spread on RGD-functionalized PEG-Nor hydrogels. MSCs encapsulated in PEG-Nor hydrogels are also highly viable, demonstrating the ability of this platform to form biocompatible hydrogels for 2D and 3D cell culture with user-defined mechanical and biochemical properties.
Collapse
Affiliation(s)
- Umu S Jalloh
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Arielle Gsell
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Kirstene A Gultian
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - James MacAulay
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Abigail Madden
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Jillian Smith
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Luke Siri
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Sebastián L Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
- Department of Orthopaedic Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| |
Collapse
|
12
|
Pishnamazi SM, Ghaderian SMH, Irani S, Ardeshirylajimi A. Polycaprolactone/poly L-lactic acid nanofibrous scaffold improves osteogenic differentiation of the amniotic fluid-derived stem cells. In Vitro Cell Dev Biol Anim 2024; 60:106-114. [PMID: 38123755 DOI: 10.1007/s11626-023-00838-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Using stem cells is one of the most important determining factors in repairing lesions using regenerative medicine. Obtaining adult stem cells from patients is a perfect choice, but it is worth noting that their differentiation and proliferation potential decreases as the patient ages. For this reason, the use of amniotic fluid stem cells can be one of the excellent alternatives. This research aimed to investigate the osteogenic differentiation potential of the amniotic fluid stem cells while cultured on the polycaprolactone/poly L-lactic acid nanofibrous scaffold. Scaffolds were qualitatively evaluated by a scanning electron microscope, and their hydrophilicity and mechanical properties were studied using contact angle and tensile test, respectively. The biocompatibility and non-toxicity of the nanofibers were also evaluated using viability assay. The osteo-supportive capacity of the nanofibers was examined using alizarin red staining, alkaline phosphatase activity, and calcium release measurement. Finally, the expression level of four important bone-related genes was determined quantitatively. The results demonstrated that the mineralization rate, alkaline phosphatase activity, intracellular calcium, and bone-related genes increased significantly in the cells cultured on the polycaprolactone/poly L-lactic acid scaffold compared to the cells cultured on the tissue culture plate as a control. According to the results, it can be concluded that the polycaprolactone/poly L-lactic acid nanofibrous scaffold surprisingly improved the osteogenic differentiation potential of the amniotic fluid stem cells and, in combination with polycaprolactone/poly L-lactic acid nanofibers could be a promising candidate as bone implants.
Collapse
Affiliation(s)
| | | | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
13
|
Joseph JP, Gugulothu SB, Nandi D, Chatterjee K. Mechanical Properties Affect Primary T Cell Activation in 3D Bioprinted Hydrogels. ACS Macro Lett 2023; 12:1085-1093. [PMID: 37466277 DOI: 10.1021/acsmacrolett.3c00271] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
T cells play a critical role in the adaptive immune response of the body, especially against intracellular pathogens and cancer. In vitro, T cell activation studies typically employ planar (two-dimensional, 2D) culture systems that do not mimic native cell-to-extracellular matrix (ECM) interactions, which influence activation. The goal of this work was to study T cell responses in a cell line (EL4) and primary mouse T cells in three-dimensional (3D) bioprinted matrices of varied stiffness. Cell-laden hydrogels were 3D bioprinted from gelatin methacryloyl (GelMA) using a digital light processing (DLP)-based 3D bioprinter operated with visible light (405 nm). Mechanical characterization revealed that the hydrogels had pathophysiologically relevant stiffnesses for a lymph node-mimetic tissue construct. EL4, a mouse T cell lymphoma line, or primary mouse T cells were 3D bioprinted and activated using a combination of 10 ng/mL of phorbol myristate acetate (PMA) and 0.1 μM of ionomycin. Cellular responses revealed differences between 2D and 3D cultures and that the biomechanical properties of the 3D bioprinted hydrogel influence T cell activation. Cellular responses of the 2D and 3D cultures in a soft matrix (19.83 ± 2.36 kPa) were comparable; however, they differed in a stiff matrix (52.95 ± 1.36 kPa). The fraction of viable EL4 cells was 1.3-fold higher in the soft matrix than in the stiff matrix. Furthermore, primary mouse T cells activated with PMA and ionomycin showed 1.35-fold higher viable cells in the soft matrix than in the stiff matrix. T cells bioprinted in a soft matrix and a stiff matrix released 7.4-fold and 5.9-fold higher amounts of interleukin-2 (IL-2) than 2D cultured cells, respectively. Overall, the study demonstrates the changes in the response of T cells in 3D bioprinted scaffolds toward engineering an ex vivo lymphoid tissue-mimetic system that can faithfully recapitulate T cell activation and unravel pathophysiological characteristics of T cells in infectious biology, autoimmunity, and cancers.
Collapse
Affiliation(s)
- Joel P Joseph
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru - 560012, India
| | | | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bengaluru - 560012, India
| | - Kaushik Chatterjee
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru - 560012, India
- Department of Materials Engineering, Indian Institute of Science, Bengaluru - 560012, India
| |
Collapse
|
14
|
Mukherjee S, Kim B, Cheng LY, Doerfert MD, Li J, Hernandez A, Liang L, Jarvis MI, Rios PD, Ghani S, Joshi I, Isa D, Ray T, Terlier T, Fell C, Song P, Miranda RN, Oberholzer J, Zhang DY, Veiseh O. Screening hydrogels for antifibrotic properties by implanting cellularly barcoded alginates in mice and a non-human primate. Nat Biomed Eng 2023; 7:867-886. [PMID: 37106151 PMCID: PMC10593184 DOI: 10.1038/s41551-023-01016-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 02/27/2023] [Indexed: 04/29/2023]
Abstract
Screening implantable biomaterials for antifibrotic properties is constrained by the need for in vivo testing. Here we show that the throughput of in vivo screening can be increased by cellularly barcoding a chemically modified combinatorial library of hydrogel formulations. The method involves the implantation of a mixture of alginate formulations, each barcoded with human umbilical vein endothelial cells from different donors, and the association of the identity and performance of each formulation by genotyping single nucleotide polymorphisms of the cells via next-generation sequencing. We used the method to screen 20 alginate formulations in a single mouse and 100 alginate formulations in a single non-human primate, and identified three lead hydrogel formulations with antifibrotic properties. Encapsulating human islets with one of the formulations led to long-term glycaemic control in a mouse model of diabetes, and coating medical-grade catheters with the other two formulations prevented fibrotic overgrowth. High-throughput screening of barcoded biomaterials in vivo may help identify formulations that enhance the long-term performance of medical devices and of biomaterial-encapsulated therapeutic cells.
Collapse
Affiliation(s)
- Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, TX, USA
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Lauren Y Cheng
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Jiaming Li
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Lily Liang
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Maria I Jarvis
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | | | | | | | - Trisha Ray
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Tanguy Terlier
- SIMS Laboratory, Shared Equipment Authority, Rice University, Houston, TX, USA
| | - Cody Fell
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Ping Song
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Roberto N Miranda
- Department of Hematopathology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jose Oberholzer
- Division of Transplant Surgery, University of Virginia, Charlottesville, VA, USA
| | - David Yu Zhang
- Department of Bioengineering, Rice University, Houston, TX, USA.
- NuProbe USA, Houston, TX, USA.
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
15
|
Lee H, Shin DY, Na Y, Han G, Kim J, Kim N, Bang SJ, Kang HS, Oh S, Yoon CB, Park J, Kim HE, Jung HD, Kang MH. Antibacterial PLA/Mg composite with enhanced mechanical and biological performance for biodegradable orthopedic implants. BIOMATERIALS ADVANCES 2023; 152:213523. [PMID: 37336010 DOI: 10.1016/j.bioadv.2023.213523] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/29/2023] [Accepted: 06/12/2023] [Indexed: 06/21/2023]
Abstract
Biodegradability, bone-healing rate, and prevention of bacterial infection are critical factors for orthopedic implants. Polylactic acid (PLA) is a good candidate biodegradable material; however, it has insufficient mechanical strength and bioactivity for orthopedic implants. Magnesium (Mg), has good bioactivity, biodegradability, and sufficient mechanical properties, similar to that of bone. Moreover, Mg has an inherent antibacterial property via a photothermal effect, which generates localized heat, thus preventing bacterial infection. Therefore, Mg is a good candidate material for PLA composites, to improve their mechanical and biological performance and add an antibacterial property. Herein, we fabricated an antibacterial PLA/Mg composite for enhanced mechanical and biological performance with an antibacterial property for application as biodegradable orthopedic implants. The composite was fabricated with 15 and 30 vol% of Mg homogeneously dispersed in PLA without the generation of a defect using a high-shear mixer. The composites exhibited an enhanced compressive strength of 107.3 and 93.2 MPa, and stiffness of 2.3 and 2.5 GPa, respectively, compared with those of pure PLA which were 68.8 MPa and 1.6 GPa, respectively. Moreover, the PLA/Mg composite at 15 vol% Mg exhibited significant improvement of biological performance in terms of enhanced initial cell attachment and cell proliferation, whereas the composite at 30 vol% Mg showed deteriorated cell proliferation and differentiation because of the rapid degradation of the Mg particles. In turn, the PLA/Mg composites exerted an antibacterial effect based on the inherent antibacterial property of Mg as well as the photothermal effect induced by near-infrared (NIR) treatment, which can minimize infection after implantation surgery. Therefore, antibacterial PLA/Mg composites with enhanced mechanical and biological performance may be a candidate material with great potential for biodegradable orthopedic implants.
Collapse
Affiliation(s)
- Hyun Lee
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Da-Young Shin
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yuhyun Na
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Ginam Han
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Joodeok Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Nahyun Kim
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Seo-Jun Bang
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Hyeong Seok Kang
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - SeKwon Oh
- Research Institute of Advanced Manufacturing & Materials Technology, Korea Institute of Industrial Technology, Incheon 21999, Republic of Korea
| | - Chang-Bun Yoon
- Department of Advanced Materials Engineering, Tech University of Korea, Siheung-si 15073, Republic of Korea
| | - Jungwon Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea; Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul 08826, Republic of Korea; Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea; Advanced Institutes of Convergence Technology, Seoul National University, Suwon-si 16229, Republic of Korea
| | - Hyoun-Ee Kim
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun-Do Jung
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Min-Ho Kang
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea.
| |
Collapse
|
16
|
Shehzad A, Mukasheva F, Moazzam M, Sultanova D, Abdikhan B, Trifonov A, Akilbekova D. Dual-Crosslinking of Gelatin-Based Hydrogels: Promising Compositions for a 3D Printed Organotypic Bone Model. Bioengineering (Basel) 2023; 10:704. [PMID: 37370635 DOI: 10.3390/bioengineering10060704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Gelatin-based hydrogels have emerged as a popular scaffold material for tissue engineering applications. The introduction of variable crosslinking methods has shown promise for fabricating stable cell-laden scaffolds. In this work, we examine promising composite biopolymer-based inks for extrusion-based 3D bioprinting, using a dual crosslinking approach. A combination of carefully selected printable hydrogel ink compositions and the use of photoinduced covalent and ionic crosslinking mechanisms allows for the fabrication of scaffolds of high accuracy and low cytotoxicity, resulting in unimpeded cell proliferation, extracellular matrix deposition, and mineralization. Three selected bioink compositions were characterized and the respective cell-laden scaffolds were bioprinted. Temporal stability, morphology, swelling, and mechanical properties of the scaffolds were thoroughly studied and the biocompatibility of the constructs was assessed using rat mesenchymal stem cells while focusing on osteogenesis. Experimental results showed that the composition of 1% alginate, 4% gelatin, and 5% (w/v) gelatine methacrylate, was found to be optimal among the examined, with shape fidelity of 88%, large cell spreading area and cell viability at around 100% after 14 days. The large pore diameters that exceed 100 µm, and highly interconnected scaffold morphology, make these hydrogels extremely potent in bone tissue engineering and bone organoid fabrication.
Collapse
Affiliation(s)
- Ahmer Shehzad
- Department of Chemical Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Fariza Mukasheva
- Department of Chemical Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Muhammad Moazzam
- Department of Chemical Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Dana Sultanova
- Department of Chemical Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Birzhan Abdikhan
- Department of Chemical Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Alexander Trifonov
- Department of Chemical Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Dana Akilbekova
- Department of Chemical Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
17
|
Shou Y, Liu L, Liu Q, Le Z, Lee KL, Li H, Li X, Koh DZ, Wang Y, Liu TM, Yang Z, Lim CT, Cheung C, Tay A. Mechano-responsive hydrogel for direct stem cell manufacturing to therapy. Bioact Mater 2023; 24:387-400. [PMID: 36632503 PMCID: PMC9817177 DOI: 10.1016/j.bioactmat.2022.12.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 01/04/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cell (MSC) is one of the most actively studied cell types due to its regenerative potential and immunomodulatory properties. Conventional cell expansion methods using 2D tissue culture plates and 2.5D microcarriers in bioreactors can generate large cell numbers, but they compromise stem cell potency and lack mechanical preconditioning to prepare MSC for physiological loading expected in vivo. To overcome these challenges, in this work, we describe a 3D dynamic hydrogel using magneto-stimulation for direct MSC manufacturing to therapy. With our technology, we found that dynamic mechanical stimulation (DMS) enhanced matrix-integrin β1 interactions which induced MSCs spreading and proliferation. In addition, DMS could modulate MSC biofunctions including directing MSC differentiation into specific lineages and boosting paracrine activities (e.g., growth factor secretion) through YAP nuclear localization and FAK-ERK pathway. With our magnetic hydrogel, complex procedures from MSC manufacturing to final clinical use, can be integrated into one single platform, and we believe this 'all-in-one' technology could offer a paradigm shift to existing standards in MSC therapy.
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Ling Liu
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| | - Qimin Liu
- School of Civil Engineering and Architecture, Wuhan University of Technology, 430070, Wuhan, China
| | - Zhicheng Le
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Khang Leng Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
| | - Hua Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 639798, Singapore
| | - Xianlei Li
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Dion Zhanyun Koh
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Yuwen Wang
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Tong Ming Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Zheng Yang
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119288, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| |
Collapse
|
18
|
Liu J, Yang L, Liu K, Gao F. Hydrogel scaffolds in bone regeneration: Their promising roles in angiogenesis. Front Pharmacol 2023; 14:1050954. [PMID: 36860296 PMCID: PMC9968752 DOI: 10.3389/fphar.2023.1050954] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Bone tissue engineering (BTE) has become a hopeful potential treatment strategy for large bone defects, including bone tumors, trauma, and extensive fractures, where the self-healing property of bone cannot repair the defect. Bone tissue engineering is composed of three main elements: progenitor/stem cells, scaffold, and growth factors/biochemical cues. Among the various biomaterial scaffolds, hydrogels are broadly used in bone tissue engineering owing to their biocompatibility, controllable mechanical characteristics, osteoconductive, and osteoinductive properties. During bone tissue engineering, angiogenesis plays a central role in the failure or success of bone reconstruction via discarding wastes and providing oxygen, minerals, nutrients, and growth factors to the injured microenvironment. This review presents an overview of bone tissue engineering and its requirements, hydrogel structure and characterization, the applications of hydrogels in bone regeneration, and the promising roles of hydrogels in bone angiogenesis during bone tissue engineering.
Collapse
Affiliation(s)
- Jun Liu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Lili Yang
- Department of Spinal Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Kexin Liu
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Feng Gao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China,*Correspondence: Feng Gao,
| |
Collapse
|
19
|
Mavinga M, Palmier M, Rémy M, Jeannière C, Lenoir S, Rey S, Saint-Marc M, Alonso F, Génot E, Thébaud N, Chevret E, Mournetas V, Rousseau B, Boiziau C, Boeuf H. The Journey of SCAPs (Stem Cells from Apical Papilla), from Their Native Tissue to Grafting: Impact of Oxygen Concentration. Cells 2022; 11:cells11244098. [PMID: 36552862 PMCID: PMC9776846 DOI: 10.3390/cells11244098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Tissue engineering strategies aim at characterizing and at optimizing the cellular component that is combined with biomaterials, for improved tissue regeneration. Here, we present the immunoMap of apical papilla, the native tissue from which SCAPs are derived. We characterized stem cell niches that correspond to a minority population of cells expressing Mesenchymal stromal/Stem Cell (CD90, CD105, CD146) and stemness (SSEA4 and CD49f) markers as well as endothelial cell markers (VWF, CD31). Based on the colocalization of TKS5 and cortactin markers, we detected migration-associated organelles, podosomes-like structures, in specific regions and, for the first time, in association with stem cell niches in normal tissue. From six healthy teenager volunteers, each with two teeth, we derived twelve cell banks, isolated and amplified under 21 or 3% O2. We confirmed a proliferative advantage of all banks when cultured under 3% versus 21% O2. Interestingly, telomerase activity was similar to that of the highly proliferative hiPSC cell line, but unrelated to O2 concentration. Finally, SCAPs embedded in a thixotropic hydrogel and implanted subcutaneously in immunodeficient mice were protected from cell death with a slightly greater advantage for cells preconditioned at 3% O2.
Collapse
Affiliation(s)
- Marine Mavinga
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | | | - Murielle Rémy
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | | | - Solène Lenoir
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | - Sylvie Rey
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | | | - Florian Alonso
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | - Elisabeth Génot
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | - Noélie Thébaud
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | - Edith Chevret
- Univ. Bordeaux, INSERM, BRIC, U1312, F-33000 Bordeaux, France
| | | | - Benoit Rousseau
- Univ. Bordeaux, Animal Facility A2, Service Commun des Animaleries, F-33000 Bordeaux, France
| | | | - Helene Boeuf
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
20
|
Chen A, Tian H, Yang N, Zhang Z, Yang GY, Cui W, Tang Y. Towards extracellular vesicle delivery systems for tissue regeneration: material design at the molecular level. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:323-356. [PMID: 39697358 PMCID: PMC11648451 DOI: 10.20517/evcna.2022.37] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 12/20/2024]
Abstract
The discovery and development of extracellular vesicles in tissue engineering have shown great potential for tissue regenerative therapies. However, their vesicle nature requires dosage-dependent administration and efficient interactions with recipient cells. Researchers have resorted to biomaterials for localized and sustained delivery of extracellular vesicles to the targeted cells, but not much emphasis has been paid on the design of the materials, which deeply impacts their molecular interactions with the loaded extracellular vesicles and subsequent delivery. Therefore, we present in this review a comprehensive survey of extracellular vesicle delivery systems from the viewpoint of material design at the molecular level. We start with general requirements of the materials and delve into different properties of delivery systems as a result of different designs, from material selections to processing strategies. Based on these differences, we analyzed the performance of extracellular vesicle delivery and tissue regeneration in representative studies. In light of the current missing links within the relationship of material structures, physicochemical properties and delivery performances, we provide perspectives on the interactions of materials and extracellular vesicles and the possible extension of materials. This review aims to be a strategic enlightenment for the future design of extracellular vesicle delivery systems to facilitate their translation from basic science to clinical applications.
Collapse
Affiliation(s)
- Ao Chen
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hengli Tian
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Nana Yang
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, Shandong, China
| | - Zhijun Zhang
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yaohui Tang
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
21
|
Martinez-Garcia FD, Fischer T, Hayn A, Mierke CT, Burgess JK, Harmsen MC. A Beginner’s Guide to the Characterization of Hydrogel Microarchitecture for Cellular Applications. Gels 2022; 8:gels8090535. [PMID: 36135247 PMCID: PMC9498492 DOI: 10.3390/gels8090535] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a three-dimensional, acellular scaffold of living tissues. Incorporating the ECM into cell culture models is a goal of cell biology studies and requires biocompatible materials that can mimic the ECM. Among such materials are hydrogels: polymeric networks that derive most of their mass from water. With the tuning of their properties, these polymer networks can resemble living tissues. The microarchitectural properties of hydrogels, such as porosity, pore size, fiber length, and surface topology can determine cell plasticity. The adequate characterization of these parameters requires reliable and reproducible methods. However, most methods were historically standardized using other biological specimens, such as 2D cell cultures, biopsies, or even animal models. Therefore, their translation comes with technical limitations when applied to hydrogel-based cell culture systems. In our current work, we have reviewed the most common techniques employed in the characterization of hydrogel microarchitectures. Our review provides a concise description of the underlying principles of each method and summarizes the collective data obtained from cell-free and cell-loaded hydrogels. The advantages and limitations of each technique are discussed, and comparisons are made. The information presented in our current work will be of interest to researchers who employ hydrogels as platforms for cell culture, 3D bioprinting, and other fields within hydrogel-based research.
Collapse
Affiliation(s)
- Francisco Drusso Martinez-Garcia
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands
- W.J. Kolff Research Institute, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Tony Fischer
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Science, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
| | - Alexander Hayn
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Science, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
- Clinic and Polyclinic for Oncology, Gastroenterology, Hepatology, Pneumology, Infectiology Department of Hepatology, University Hospital Leipzig, Liebigstr. 19, 04103 Leipzig, Germany
| | - Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Science, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
- Correspondence: (C.T.M.); (M.C.H.)
| | - Janette Kay Burgess
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands
- W.J. Kolff Research Institute, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 AV Groningen, The Netherlands
| | - Martin Conrad Harmsen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands
- W.J. Kolff Research Institute, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 AV Groningen, The Netherlands
- Correspondence: (C.T.M.); (M.C.H.)
| |
Collapse
|
22
|
Hassani A, Avci ÇB, Kerdar SN, Amini H, Amini M, Ahmadi M, Sakai S, Bagca BG, Ozates NP, Rahbarghazi R, Khoshfetrat AB. Interaction of alginate with nano-hydroxyapatite-collagen using strontium provides suitable osteogenic platform. J Nanobiotechnology 2022; 20:310. [PMID: 35765003 PMCID: PMC9238039 DOI: 10.1186/s12951-022-01511-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/14/2022] [Indexed: 11/10/2022] Open
Abstract
Background Hydrogels based on organic/inorganic composites have been at the center of attention for the fabrication of engineered bone constructs. The establishment of a straightforward 3D microenvironment is critical to maintaining cell-to-cell interaction and cellular function, leading to appropriate regeneration. Ionic cross-linkers, Ca2+, Ba2+, and Sr2+, were used for the fabrication of Alginate-Nanohydroxyapatite-Collagen (Alg-nHA-Col) microspheres, and osteogenic properties of human osteoblasts were examined in in vitro and in vivo conditions after 21 days. Results Physicochemical properties of hydrogels illustrated that microspheres cross-linked with Sr2+ had reduced swelling, enhanced stability, and mechanical strength, as compared to the other groups. Human MG-63 osteoblasts inside Sr2+ cross-linked microspheres exhibited enhanced viability and osteogenic capacity indicated by mineralization and the increase of relevant proteins related to bone formation. PCR (Polymerase Chain Reaction) array analysis of the Wnt (Wingless-related integration site) signaling pathway revealed that Sr2+ cross-linked microspheres appropriately induced various signaling transduction pathways in human osteoblasts leading to osteogenic activity and dynamic growth. Transplantation of Sr2+ cross-linked microspheres with rat osteoblasts into cranium with critical size defect in the rat model accelerated bone formation analyzed with micro-CT and histological examination. Conclusion Sr2+ cross-linked Alg-nHA-Col hydrogel can promote functionality and dynamic growth of osteoblasts. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01511-9.
Collapse
Affiliation(s)
- Ayla Hassani
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, 51335-1996, Iran.,Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Çığır Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Sajed Nazif Kerdar
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, 51335-1996, Iran.,Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Hassan Amini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Meisam Amini
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
| | - Mahdi Ahmadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shinji Sakai
- Division of Chemical Engineering, Department of Materials Science and Engineering, Graduate School of Engineering Science, Osaka University, Osaka, 560-8531, Japan
| | - Bakiye Goker Bagca
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | | | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ali Baradar Khoshfetrat
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, 51335-1996, Iran. .,Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran.
| |
Collapse
|
23
|
Chen Y, Lin J, Yan W. A Prosperous Application of Hydrogels With Extracellular Vesicles Release for Traumatic Brain Injury. Front Neurol 2022; 13:908468. [PMID: 35720072 PMCID: PMC9201053 DOI: 10.3389/fneur.2022.908468] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/05/2022] [Indexed: 01/29/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of disability worldwide, becoming a heavy burden to the family and society. However, the complexity of the brain and the existence of blood-brain barrier (BBB) do limit most therapeutics effects through simple intravascular injection. Hence, an effective therapy promoting neurological recovery is urgently required. Although limited spontaneous recovery of function post-TBI does occur, increasing evidence indicates that exosomes derived from stem cells promote these endogenous processes. The advantages of hydrogels for transporting drugs and stem cells to target injured sites have been discussed in multitudinous studies. Therefore, the combined employment of hydrogels and exosomes for TBI is worthy of further study. Herein, we review current research associated with the application of hydrogels and exosomes for TBI. We also discuss the possibilities and advantages of exosomes and hydrogels co-therapies after TBI.
Collapse
|
24
|
Preobrazhenskii II, Putlyaev VI. 3D Printing of Hydrogel-Based Biocompatible Materials. RUSS J APPL CHEM+ 2022. [DOI: 10.1134/s1070427222060027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
25
|
Im S, Choe G, Seok JM, Yeo SJ, Lee JH, Kim WD, Lee JY, Park SA. An osteogenic bioink composed of alginate, cellulose nanofibrils, and polydopamine nanoparticles for 3D bioprinting and bone tissue engineering. Int J Biol Macromol 2022; 205:520-529. [PMID: 35217077 DOI: 10.1016/j.ijbiomac.2022.02.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/20/2022] [Accepted: 02/05/2022] [Indexed: 12/16/2022]
Abstract
Bioprinting is an emerging technology for manufacturing cell-laden three-dimensional (3D) scaffolds, which are used to fabricate complex 3D constructs and provide specific microenvironments for supporting cell growth and differentiation. The development of bioinks with appropriate printability and specific bioactivities is crucial for bioprinting and tissue engineering applications, including bone tissue regeneration. Therefore, to produce functional bioinks for osteoblast printing and bone tissue formation, we formulated various nanocomposite hydrogel-based bioinks using natural and biocompatible biomaterials (i.e., alginate, tempo-oxidized cellulose nanofibrils (TOCNF), and polydopamine nanoparticles (PDANPs)). Rheological studies and printability tests revealed that bioinks containing 1.5% alginate and 1.5% TOCNF in the presence or absence of PDANP (0.5%) are suitable for 3D printing. Furthermore, in vitro studies of 3D-printed osteoblast-laden scaffolds indicated that the 0.5% PDANP-incorporated bioink induced significant osteogenesis. Overall, the bioink consisting of alginate, TOCNF, and PDANPs exhibited excellent printability and bioactivity (i.e., osteogenesis).
Collapse
Affiliation(s)
- Seunghyun Im
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), 156 Gajeongbuk-ro, Yuseong-gu, Daejeon 304-343, Republic of Korea; School of Materials Science and Engineering, Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 500-712, Republic of Korea
| | - Goeun Choe
- School of Materials Science and Engineering, Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 500-712, Republic of Korea
| | - Ji Min Seok
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), 156 Gajeongbuk-ro, Yuseong-gu, Daejeon 304-343, Republic of Korea
| | - Seon Ju Yeo
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), 156 Gajeongbuk-ro, Yuseong-gu, Daejeon 304-343, Republic of Korea
| | - Jun Hee Lee
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), 156 Gajeongbuk-ro, Yuseong-gu, Daejeon 304-343, Republic of Korea
| | - Wan Doo Kim
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), 156 Gajeongbuk-ro, Yuseong-gu, Daejeon 304-343, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 500-712, Republic of Korea
| | - Su A Park
- Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery and Materials (KIMM), 156 Gajeongbuk-ro, Yuseong-gu, Daejeon 304-343, Republic of Korea.
| |
Collapse
|
26
|
Assessment of Collagen-Based Nanostructured Biomimetic Systems with a Co-Culture of Human Bone-Derived Cells. Cells 2021; 11:cells11010026. [PMID: 35011588 PMCID: PMC8744918 DOI: 10.3390/cells11010026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/03/2021] [Accepted: 12/14/2021] [Indexed: 12/25/2022] Open
Abstract
Osteoporosis is a worldwide disease resulting in the increase of bone fragility and enhanced fracture risk in adults. In the context of osteoporotic fractures, bone tissue engineering (BTE), i.e., the use of bone substitutes combining biomaterials, cells, and other factors, is considered a potential alternative to conventional treatments. Innovative scaffolds need to be tested in in vitro systems where the simultaneous presence of osteoblasts (OBs) and osteoclasts (OCs), the two main players of bone remodeling, is required to mimic their crosstalk and molecular cooperation. To this aim, two composite materials were developed, based on type I collagen, and containing either strontium-enriched mesoporous bioactive glasses or rod-like hydroxyapatite nanoparticles. The developed nanostructured systems underwent genipin chemical crosslinking and were then tested with an indirect co-culture of human trabecular bone-derived OBs and buffy coat-derived OC precursors, for 2–3 weeks. The favorable structural and biological properties of the materials proved to successfully support the viability, adhesion, and differentiation of cells, encouraging a further investigation of the developed bioactive systems as biomaterial inks for the 3D printing of more complex scaffolds for BTE.
Collapse
|
27
|
Wu M, Zhao Y, Jiang H, Xu X, Wang D, Xu X, Zhou Y, Tan H, Ding C, Li J. Self-Organized Spatiotemporal Mineralization of Hydrogel: A Simulant of Osteon. SMALL 2021; 18:e2106649. [PMID: 34921591 DOI: 10.1002/smll.202106649] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/27/2021] [Indexed: 02/05/2023]
Abstract
Nature creates fascinating self-organized spatiotemporal patterns through the delicate control of reaction-diffusion dynamics. As the primary unit of cortical bone, osteon has concentric lamellar architecture, which plays a crucial role in the mechanical and physiological functions of bone. However, it remains a great challenge to fabricate the osteon-like structure in a natural self-organization way. Taking advantage of the nonequilibrium reaction in hydrogels, a simple mineralization strategy to closely mimic the formation of osteon in a mild physiological condition is developed. By constructing two reverse concentration gradients of ions from periphery to interior of cylindrical hydrogel, spatiotemporal self-organization of calcium phosphate in concentric rings is generated. It is noteworthy that minerals in different layers possess diverse contents and crystalline phases, which further guide the adhesion and spread of osteoblasts on these patterns, resembling the architecture and cytological behavior of osteon. Besides, theoretical data indicates the predominate role of ion concentrations and pH values of solution, in good accordance with experimental results. Independent of precise instruments, this lifelike method is easily obtained, cost-efficient, and effectively imitates the mineral deposition in osteon from a physiochemical view. The strategy may be expanded to develop other functional material patterns via spatiotemporal self-organization.
Collapse
Affiliation(s)
- Mingzhen Wu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Yao Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Haolun Jiang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xiaoyang Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Dingqian Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xinyuan Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Yahong Zhou
- 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
| | - 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
| | - 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, Chengdu, 610041, China
| |
Collapse
|
28
|
Nazir F, Abbas L, Iqbal M. A comparative insight into the mechanical properties, antibacterial potential, and cytotoxicity profile of nano-hydroxyapatite and nano-whitlockite-incorporated poly-L-lactic acid for bone tissue engineering. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-021-02223-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
29
|
Harris AF, Lacombe J, Zenhausern F. The Emerging Role of Decellularized Plant-Based Scaffolds as a New Biomaterial. Int J Mol Sci 2021; 22:12347. [PMID: 34830229 PMCID: PMC8625747 DOI: 10.3390/ijms222212347] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/30/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022] Open
Abstract
The decellularization of plant-based biomaterials to generate tissue-engineered substitutes or in vitro cellular models has significantly increased in recent years. These vegetal tissues can be sourced from plant leaves and stems or fruits and vegetables, making them a low-cost, accessible, and sustainable resource from which to generate three-dimensional scaffolds. Each construct is distinct, representing a wide range of architectural and mechanical properties as well as innate vasculature networks. Based on the rapid rise in interest, this review aims to detail the current state of the art and presents the future challenges and perspectives of these unique biomaterials. First, we consider the different existing decellularization techniques, including chemical, detergent-free, enzymatic, and supercritical fluid approaches that are used to generate such scaffolds and examine how these protocols can be selected based on plant cellularity. We next examine strategies for cell seeding onto the plant-derived constructs and the importance of the different functionalization methods used to assist in cell adhesion and promote cell viability. Finally, we discuss how their structural features, such as inherent vasculature, porosity, morphology, and mechanical properties (i.e., stiffness, elasticity, etc.) position plant-based scaffolds as a unique biomaterial and drive their use for specific downstream applications. The main challenges in the field are presented throughout the discussion, and future directions are proposed to help improve the development and use of vegetal constructs in biomedical research.
Collapse
Affiliation(s)
- Ashlee F. Harris
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA;
| | - Jerome Lacombe
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA;
- Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA
| | - Frederic Zenhausern
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA;
- Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
30
|
Hydrogel, Electrospun and Composite Materials for Bone/Cartilage and Neural Tissue Engineering. MATERIALS 2021; 14:ma14226899. [PMID: 34832300 PMCID: PMC8624846 DOI: 10.3390/ma14226899] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022]
Abstract
Injuries of the bone/cartilage and central nervous system are still a serious socio-economic problem. They are an effect of diversified, difficult-to-access tissue structures as well as complex regeneration mechanisms. Currently, commercially available materials partially solve this problem, but they do not fulfill all of the bone/cartilage and neural tissue engineering requirements such as mechanical properties, biochemical cues or adequate biodegradation. There are still many things to do to provide complete restoration of injured tissues. Recent reports in bone/cartilage and neural tissue engineering give high hopes in designing scaffolds for complete tissue regeneration. This review thoroughly discusses the advantages and disadvantages of currently available commercial scaffolds and sheds new light on the designing of novel polymeric scaffolds composed of hydrogels, electrospun nanofibers, or hydrogels loaded with nano-additives.
Collapse
|
31
|
Nazir F, Iqbal M. Comparative Study of Crystallization, Mechanical Properties, and In Vitro Cytotoxicity of Nanocomposites at Low Filler Loadings of Hydroxyapatite for Bone-Tissue Engineering Based on Poly(l-lactic acid)/Cyclo Olefin Copolymer. Polymers (Basel) 2021; 13:3865. [PMID: 34833163 PMCID: PMC8619963 DOI: 10.3390/polym13223865] [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: 09/01/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 12/23/2022] Open
Abstract
A poly(l-lactic acid)/nanohydroxyapatite (PLLA/nHA) scaffold works as a bioactive, osteoconductive scaffold for bone-tissue engineering, but its low degradation rate limits embedded HA in PLLA to efficiently interact with body fluids. In this work, nano-hydroxyapatite (nHA) was added in lower filler loadings (1, 5, 10, and 20 wt%) in a poly(l-lactic acid)/cyclo olefin copolymer10 wt% (PLLA/COC10) blend to obtain novel poly(l-lactic acid)/cyclo olefin copolymer/nanohydroxyapatite (PLLA/COC10-nHA) scaffolds for bone-tissue regeneration and repair. Furthermore, the structure-activity relationship of PLLA/COC10-nHA (ternary system) nanocomposites in comparison with PLLA/nHA (binary system) nanocomposites was systematically studied. Nanocomposites were evaluated for structural (morphology, crystallization), thermomechanical properties, antibacterial potential, and cytocompatibility for bone-tissue engineering applications. Scanning electron microscope images revealed that PLLA/COC10-nHA had uniform morphology and dispersion of nanoparticles up to 10% of HA, and the overall nHA dispersion in matrix was better in PLLA/COC10-nHA as compared to PLLA/nHA. Fourier transformation infrared spectroscopy (FTIR), powder X-ray diffraction (XRD), and differential scanning calorimetry (DSC) studies confirmed miscibility and transformation of the α-crystal form of PLLA to the ά-crystal form by the addition of nHA in all nanocomposites. The degree of crystallinity (%) in the case of PLLA/COC10-nHA 10 wt% was 114% higher than pure PLLA/COC10 and 128% higher than pristine PLLA, indicating COC and nHA are acting as nucleating agents in the PLLA/COC10-nHA nanocomposites, causing an increase in the degree of crystallinity (%). Moreover, PLLA/COC10-nHA exhibited 140 to 240% (1-20 wt% HA) enhanced mechanical properties in terms of ductility as compared to PLLA/nHA. Antibacterial activity results showed that 10 wt% HA in PLLA/COC10-nHA showed substantial activity against P. aeruginosa, S. aureus, and L. monocytogenes. In vitro cytocompatibility of PLLA/COC10 and PLLA nanocomposites with nHA osteoprogenitor cells (MC3T3-E1) and bone mesenchymal stem cells (BMSC) was evaluated. Both cell lines showed two- to three-fold enhancement in cell viability and 10- to 30-fold in proliferation upon culture on PLLA/COC10-nHA as compared to PLLA/nHA composites. It was observed that the ternary system PLLA/COC10-nHA had good dispersion and interfacial interaction resulting in improved thermomechanical and enhanced osteoconductive properties as compared to PLLA/nHA.
Collapse
Affiliation(s)
| | - Mudassir Iqbal
- Department of Chemistry, School of Natural Sciences, National University of Science and Technology (NUST), Islamabad 44000, Pakistan;
| |
Collapse
|
32
|
Ender AM, Kaygisiz K, Räder HJ, Mayer FJ, Synatschke CV, Weil T. Cell-Instructive Surface Gradients of Photoresponsive Amyloid-like Fibrils. ACS Biomater Sci Eng 2021; 7:4798-4808. [PMID: 34515483 PMCID: PMC8512672 DOI: 10.1021/acsbiomaterials.1c00889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Gradients of bioactive molecules play a crucial role in various biological processes like vascularization, tissue regeneration, or cell migration. To study these complex biological systems, it is necessary to control the concentration of bioactive molecules on their substrates. Here, we created a photochemical strategy to generate gradients using amyloid-like fibrils as scaffolds functionalized with a model epitope, that is, the integrin-binding peptide RGD, to modulate cell adhesion. The self-assembling β-sheet forming peptide (CKFKFQF) was connected to the RGD epitope via a photosensitive nitrobenzyl linker and assembled into photoresponsive nanofibrils. The fibrils were spray-coated on glass substrates and macroscopic gradients were generated by UV-light over a centimeter-scale. We confirmed the gradient formation using matrix-assisted laser desorption ionization mass spectroscopy imaging (MALDI-MSI), which directly visualizes the molecular species on the surface. The RGD gradient was used to instruct cells. In consequence, A549 adapted their adhesion properties in dependence of the RGD-epitope density.
Collapse
Affiliation(s)
- Adriana Maria Ender
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Kübra Kaygisiz
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Hans-Joachim Räder
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Franz J Mayer
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Christopher V Synatschke
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Tanja Weil
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
33
|
Kahl M, Schneidereit D, Bock N, Friedrich O, Hutmacher DW, Meinert C. MechAnalyze: An Algorithm for Standardization and Automation of Compression Test Analysis. Tissue Eng Part C Methods 2021; 27:529-542. [PMID: 34541882 DOI: 10.1089/ten.tec.2021.0170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The mechanical properties of hydrogels, as well as native and engineered tissues are key parameters frequently assessed in biomaterial science and tissue engineering research. However, a lack of standardized methods and user-independent data analysis has impacted the research community for many decades and contributes to poor reproducibility and comparability of datasets, representing a significant issue often neglected in publications. In this study, we provide a software package, MechAnalyze, facilitating the standardized and automated analysis of force-displacement data generated in unconfined compression tests. Using comparative studies of datasets analyzed manually and with MechAnalyze, we demonstrate that the software reliably determines the compressive moduli, failure stress and failure strain of hydrogels, as well as engineered and native tissues, while providing an intuitive user interface that requires minimal user input. MechAnalyze provides a fast and user-independent data analysis method and advances process standardization, reproducibility, and comparability of data for the mechanical characterization of biomaterials as well as native and engineered tissues. Impact statement Mechanical properties of hydrogels are crucial parameters in the development of new materials for tissue engineering. However, manual assessment is tedious, not standardized and suffers under user-to-user bias. Hence, the here presented stand-alone software package provides analysis and statistics of force-displacement and material geometry data to determine the compressive moduli, failure stress, and failure strain in a standardized, robust, and automated fashion. MechAnalyze will substantially support biomechanical testing of hydrogels as well as engineered and native tissues and will thus, be of appreciable value to a broad target group in regenerative medicine, tissue engineering, but also life sciences and biomedicine.
Collapse
Affiliation(s)
- Melanie Kahl
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany.,Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Dominik Schneidereit
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Nathalie Bock
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Queensland University of Technology, Woolloongabba, Australia
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany.,Centre for Biomedical Technologies, Science and Engineering Faculty, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
| | - Dietmar W Hutmacher
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia.,Centre for Biomedical Technologies, Science and Engineering Faculty, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia.,ARC ITTC in Additive Biomanufacturing, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology, Brisbane, Australia
| | - Christoph Meinert
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Centre for Biomedical Technologies, Science and Engineering Faculty, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia.,Herston Biofabrication Institute, Metro North Hospital and Health Services, Brisbane, Australia
| |
Collapse
|
34
|
Singh M, Zhang J, Bethel K, Liu Y, Davis EM, Zeng H, Kong Z, Johnson BN. Closed-Loop Controlled Photopolymerization of Hydrogels. ACS APPLIED MATERIALS & INTERFACES 2021; 13:40365-40378. [PMID: 34415733 DOI: 10.1021/acsami.1c11779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Here, we present a closed-loop controlled photopolymerization process for fabrication of hydrogels with controlled storage moduli. Hydrogel crosslinking was associated with a significant change in the phase angle of a piezoelectric cantilever sensor and established the timescale of the photopolymerization process. The composition, structure, and mechanical properties of the fabricated hydrogels were characterized using Raman spectroscopy, scanning electron microscopy (SEM), and dynamic mechanical analysis (DMA). We found that the storage moduli of photocured poly(ethylene glycol) dimethacrylate (PEGDMA) and poly(N-isopropylacrylamide) (PNIPAm) hydrogels could be controlled using bang-bang and fuzzy logic controllers. Bang-bang controlled photopolymerization resulted in constant overshoot of the storage modulus setpoint for PEGDMA hydrogels, which was mitigated by setpoint correction and fuzzy logic control. SEM and DMA studies showed that the network structure and storage modulus of PEGDMA hydrogels were dependent on the cure time and temporal profile of UV exposure during photopolymerization. This work provides an advance in pulsed and continuous photopolymerization processes for hydrogel engineering based on closed-loop control that enables reproducible fabrication of hydrogels with controlled mechanical properties.
Collapse
Affiliation(s)
- Manjot Singh
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Junru Zhang
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Keturah Bethel
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Yang Liu
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Eric M Davis
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Haibo Zeng
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Zhenyu Kong
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Blake N Johnson
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
- Department of Materials Science and Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
35
|
Soheilmoghaddam F, Rumble M, Cooper-White J. High-Throughput Routes to Biomaterials Discovery. Chem Rev 2021; 121:10792-10864. [PMID: 34213880 DOI: 10.1021/acs.chemrev.0c01026] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many existing clinical treatments are limited in their ability to completely restore decreased or lost tissue and organ function, an unenviable situation only further exacerbated by a globally aging population. As a result, the demand for new medical interventions has increased substantially over the past 20 years, with the burgeoning fields of gene therapy, tissue engineering, and regenerative medicine showing promise to offer solutions for full repair or replacement of damaged or aging tissues. Success in these fields, however, inherently relies on biomaterials that are engendered with the ability to provide the necessary biological cues mimicking native extracellular matrixes that support cell fate. Accelerating the development of such "directive" biomaterials requires a shift in current design practices toward those that enable rapid synthesis and characterization of polymeric materials and the coupling of these processes with techniques that enable similarly rapid quantification and optimization of the interactions between these new material systems and target cells and tissues. This manuscript reviews recent advances in combinatorial and high-throughput (HT) technologies applied to polymeric biomaterial synthesis, fabrication, and chemical, physical, and biological screening with targeted end-point applications in the fields of gene therapy, tissue engineering, and regenerative medicine. Limitations of, and future opportunities for, the further application of these research tools and methodologies are also discussed.
Collapse
Affiliation(s)
- Farhad Soheilmoghaddam
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Madeleine Rumble
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Justin Cooper-White
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| |
Collapse
|
36
|
Sugiyama H, Tsunemitsu K, Onoe H, Obata K, Sugioka K, Terakawa M. Microfabrication of cellulose nanofiber-reinforced hydrogel by multiphoton polymerization. Sci Rep 2021; 11:10892. [PMID: 34035420 PMCID: PMC8149388 DOI: 10.1038/s41598-021-90445-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanical strength of hydrogel microstructures is crucial for obtaining the desired flexibility, robustness, and biocompatibility for various applications such as cell scaffolds and soft microrobots. In this study, we demonstrate the fabrication of microstructures composed of cellulose nanofibers (CNFs) and poly(ethylene glycol) diacrylate (PEGDA) hydrogels by multiphoton polymerization. The stress of the fabricated microstructure during tensile testing increased with an increase in the CNF concentration, indicating that the mechanical strength of the microstructure was enhanced by using CNFs as fillers. Moreover, the swelling ratio of the microstructure increased with increasing CNF concentration in the PEGDA hydrogel. Our results show the potential of the technique for the microfabrication of advanced cell scaffolds and soft microrobots with the desired mechanical strength.
Collapse
Affiliation(s)
- Hiroki Sugiyama
- School of Integrated Design Engineering, Keio University, 3-14-1, Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan
| | - Kaneto Tsunemitsu
- School of Integrated Design Engineering, Keio University, 3-14-1, Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan
| | - Hiroaki Onoe
- School of Integrated Design Engineering, Keio University, 3-14-1, Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan.,Department of Mechanical Engineering, Keio University, 3-14-1, Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan
| | - Kotaro Obata
- Advanced Laser Processing Research Team, RIKEN Center for Advanced Photonics, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Koji Sugioka
- Advanced Laser Processing Research Team, RIKEN Center for Advanced Photonics, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Mitsuhiro Terakawa
- School of Integrated Design Engineering, Keio University, 3-14-1, Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan. .,Department of Electronics and Electrical Engineering, Keio University, 3-14-1, Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan.
| |
Collapse
|
37
|
Elkhoury K, Morsink M, Tahri Y, Kahn C, Cleymand F, Shin SR, Arab-Tehrany E, Sanchez-Gonzalez L. Synthesis and characterization of C2C12-laden gelatin methacryloyl (GelMA) from marine and mammalian sources. Int J Biol Macromol 2021; 183:918-926. [PMID: 33971227 DOI: 10.1016/j.ijbiomac.2021.05.040] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/15/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022]
Abstract
Gelatin methacryloyl (GelMA) is widely used for tissue engineering applications as an extracellular matrix (ECM) mimicking scaffold due to its cost-effectiveness, ease of synthesis, and high biocompatibility. GelMA is widely synthesized from porcine skin gelatin, which labors under clinical, religious, and economical restrictions. In order to overcome these limitations, GelMA can be produced from fish skin gelatin, which is eco-friendly as well. Here, we present a comparative study of the physicochemical (structural, thermal, water uptake, swelling, rheological, and mechanical) and biological (cell viability, proliferation, and spreading) properties of porcine and fish skin GelMA with low and high methacrylation degrees, before and after crosslinking, to check whether fish skin can replace porcine skin as the source of GelMA. Porcine and fish skin GelMA presented similar structural, thermal, and water uptake properties prior to crosslinking. However, subsequent to crosslinking, fish skin GelMA hydrogels exhibited a higher mass swelling ratio and a lower elastic and compressive Young's moduli than porcine skin GelMA hydrogels of similar methacrylation level. Both types of GelMA hydrogels showed great biocompatibility toward encapsulated mouse myoblast cells (C2C12), however, improved cell spreading was observed in fish skin GelMA hydrogels, and cell proliferation was only induced in low methacrylated GelMA. These results suggest that fish skin GelMA is a promising substitute for porcine skin GelMA for biomedical applications and that low methacrylated fish skin GelMA can be used as a potential scaffold for skeletal muscle tissue engineering.
Collapse
Affiliation(s)
- Kamil Elkhoury
- LIBio, Université de Lorraine, F-54000 Nancy, France; Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, United States of America.
| | - Margaretha Morsink
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, United States of America; Translational Liver Research, Department of Medical Cell BioPhysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands.
| | - Yasmina Tahri
- LIBio, Université de Lorraine, F-54000 Nancy, France
| | - Cyril Kahn
- LIBio, Université de Lorraine, F-54000 Nancy, France.
| | - Franck Cleymand
- Institut Jean Lamour, CNRS-Université de Lorraine, F-54000 Nancy, France.
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, United States of America.
| | | | | |
Collapse
|
38
|
Yang L, Pijuan-Galito S, Rho HS, Vasilevich AS, Eren AD, Ge L, Habibović P, Alexander MR, de Boer J, Carlier A, van Rijn P, Zhou Q. High-Throughput Methods in the Discovery and Study of Biomaterials and Materiobiology. Chem Rev 2021; 121:4561-4677. [PMID: 33705116 PMCID: PMC8154331 DOI: 10.1021/acs.chemrev.0c00752] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 02/07/2023]
Abstract
The complex interaction of cells with biomaterials (i.e., materiobiology) plays an increasingly pivotal role in the development of novel implants, biomedical devices, and tissue engineering scaffolds to treat diseases, aid in the restoration of bodily functions, construct healthy tissues, or regenerate diseased ones. However, the conventional approaches are incapable of screening the huge amount of potential material parameter combinations to identify the optimal cell responses and involve a combination of serendipity and many series of trial-and-error experiments. For advanced tissue engineering and regenerative medicine, highly efficient and complex bioanalysis platforms are expected to explore the complex interaction of cells with biomaterials using combinatorial approaches that offer desired complex microenvironments during healing, development, and homeostasis. In this review, we first introduce materiobiology and its high-throughput screening (HTS). Then we present an in-depth of the recent progress of 2D/3D HTS platforms (i.e., gradient and microarray) in the principle, preparation, screening for materiobiology, and combination with other advanced technologies. The Compendium for Biomaterial Transcriptomics and high content imaging, computational simulations, and their translation toward commercial and clinical uses are highlighted. In the final section, current challenges and future perspectives are discussed. High-throughput experimentation within the field of materiobiology enables the elucidation of the relationships between biomaterial properties and biological behavior and thereby serves as a potential tool for accelerating the development of high-performance biomaterials.
Collapse
Affiliation(s)
- Liangliang Yang
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sara Pijuan-Galito
- School
of Pharmacy, Biodiscovery Institute, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Hoon Suk Rho
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Aliaksei S. Vasilevich
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aysegul Dede Eren
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lu Ge
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Pamela Habibović
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Morgan R. Alexander
- School
of Pharmacy, Boots Science Building, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Jan de Boer
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aurélie Carlier
- Department
of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick van Rijn
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Qihui Zhou
- Institute
for Translational Medicine, Department of Stomatology, The Affiliated
Hospital of Qingdao University, Qingdao
University, Qingdao 266003, China
| |
Collapse
|
39
|
Zeng Z, Lam PT, Robinson ML, Del Rio-Tsonis K, Saul JM. Design and Characterization of Biomimetic Kerateine Aerogel-Electrospun Polycaprolactone Scaffolds for Retinal Cell Culture. Ann Biomed Eng 2021; 49:1633-1644. [PMID: 33825081 DOI: 10.1007/s10439-021-02756-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/18/2021] [Indexed: 01/23/2023]
Abstract
Age-related macular degeneration (AMD) is a retinal disease that affects 196 million people and causes nearly 9% of blindness worldwide. While several pharmacological approaches slow the effects of AMD, in our opinion, cell-based strategies offer the most likely path to a cure. We describe the design and initial characterization of a kerateine (obtained by reductive extraction from keratin proteins) aerogel-electrospun polycaprolactone fiber scaffold system. The scaffolds mimic key features of the choroid and the Bruch's membrane, which is the basement membrane to which the cells of the retinal pigment epithelium (RPE) attach. The scaffolds had elastic moduli of 2-7.2 MPa, a similar range as native choroid and Bruch's membrane. ARPE-19 cells attached to the polycaprolactone fibers, remained viable for one week, and proliferated to form a monolayer reminiscent of that needed for retinal repair. These constructs could serve as a model system for testing cell and/or drug treatment strategies or directing ex vivo retinal tissue formation in the treatment of AMD.
Collapse
Affiliation(s)
- Ziqian Zeng
- Department of Chemical, Paper and Biomedical Engineering, Miami University, 650 East High Street, Oxford, OH, 45056, USA
| | - Phuong T Lam
- Department of Biology, Miami University, Oxford, OH, USA, 45056
| | - Michael L Robinson
- Department of Biology, Miami University, Oxford, OH, USA, 45056.,Center for Visual Sciences at Miami University (CVSMU), Oxford, OH, USA
| | - Katia Del Rio-Tsonis
- Department of Biology, Miami University, Oxford, OH, USA, 45056.,Center for Visual Sciences at Miami University (CVSMU), Oxford, OH, USA
| | - Justin M Saul
- Department of Chemical, Paper and Biomedical Engineering, Miami University, 650 East High Street, Oxford, OH, 45056, USA.
| |
Collapse
|
40
|
Gwon Y, Park S, Kim W, Han T, Kim H, Kim J. Radially patterned transplantable biodegradable scaffolds as topographically defined contact guidance platforms for accelerating bone regeneration. J Biol Eng 2021; 15:12. [PMID: 33752709 PMCID: PMC7986475 DOI: 10.1186/s13036-021-00263-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The healing of large critical-sized bone defects remains a clinical challenge in modern orthopedic medicine. The current gold standard for treating critical-sized bone defects is autologous bone graft; however, it has critical limitations. Bone tissue engineering has been proposed as a viable alternative, not only for replacing the current standard treatment, but also for producing complete regeneration of bone tissue without complex surgical treatments or tissue transplantation. In this study, we proposed a transplantable radially patterned scaffold for bone regeneration that was defined by capillary force lithography technology using biodegradable polycaprolactone polymer. RESULTS The radially patterned transplantable biodegradable scaffolds had a radial structure aligned in a central direction. The radially aligned pattern significantly promoted the recruitment of host cells and migration of osteoblasts into the defect site. Furthermore, the transplantable scaffolds promoted regeneration of critical-sized bone defects by inducing cell migration and differentiation. CONCLUSIONS Our findings demonstrated that topographically defined radially patterned transplantable biodegradable scaffolds may have great potential for clinical application of bone tissue regeneration.
Collapse
Affiliation(s)
- Yonghyun Gwon
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sunho Park
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Woochan Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Taeseong Han
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyoseong Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jangho Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea.
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
41
|
Osteogenic differentiation of hBMSCs on porous photo-crosslinked poly(trimethylene carbonate) and nano-hydroxyapatite composites. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2021.110335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
42
|
Tejo-Otero A, Ritchie AC. Biological and mechanical evaluation of mineralized-hydrogel scaffolds for tissue engineering applications. J Biomater Appl 2021; 36:460-473. [PMID: 33596707 DOI: 10.1177/0885328221995425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chitosan and gelatin have been extensively used in tissue engineering for a wide range of different applications, such as wound healing or bone regeneration, due to their advantages: excellent biocompatibility (promoting cell adhesion and proliferation), low price and biodegradability. Nonetheless, their main drawback is that they have poor mechanical properties, consequently restricting their use in bone tissue engineering. In previous studies, both materials were cross-linked, with added calcium minerals, which led to an improvement in both mechanical and biological properties. Therefore, this study carries out a mechanical and biological characterization of mineral-hydrogel scaffolds in order to find the best compositions. Different proportions of calcium compounds (CaCO3 and CaHPO4) are used to make up between 20% and 30% of the minerals used in a mineral-hydrogel mix. This addition of minerals enhances not only the mechanical properties, but also the biological ones. On the one hand, the higher the amount of minerals added to the composition, the better the mechanical properties obtained. Additionally, as the proportion of CaCO3 in comparison with CaHPO4 rises, the mechanical properties improve. On the other hand, both cell proliferation and mineralization are improved with the addition of calcium minerals.
Collapse
Affiliation(s)
- Aitor Tejo-Otero
- Bioengineering Research Group, University of Nottingham, University Park Campus, Nottingham, UK
| | - Alastair C Ritchie
- Bioengineering Research Group, University of Nottingham, University Park Campus, Nottingham, UK
| |
Collapse
|
43
|
Redmond J, McCarthy H, Buchanan P, Levingstone TJ, Dunne NJ. Advances in biofabrication techniques for collagen-based 3D in vitro culture models for breast cancer research. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111944. [PMID: 33641930 DOI: 10.1016/j.msec.2021.111944] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/19/2022]
Abstract
Collagen is the most abundant component of the extracellular matrix (ECM), therefore it represents an ideal biomaterial for the culture of a variety of cell types. Recently, collagen-based scaffolds have shown promise as 3D culture platforms for breast cancer-based research. Two-dimensional (2D) in vitro culture models, while useful for gaining preliminary insights, are ultimately flawed as they do not adequately replicate the tumour microenvironment. As a result, they do not facilitate proper 3D cell-cell/cell-matrix interactions and often an exaggerated response to therapeutic agents occurs. The ECM plays a crucial role in the development and spread of cancer. Alterations within the ECM have a significant impact on the pathogenesis of cancer, the initiation of metastasis and ultimate progression of the disease. 3D in vitro culture models that aim to replicate the tumour microenvironment have the potential to offer a new frontier for cancer research with cell growth, morphology and genetic properties that more closely match in vivo cancers. While initial 3D in vitro culture models used in breast cancer research consisted of simple hydrogel platforms, recent advances in biofabrication techniques, including freeze-drying, electrospinning and 3D bioprinting, have enabled the fabrication of biomimetic collagen-based platforms that more closely replicate the breast cancer ECM. This review highlights the current application of collagen-based scaffolds as 3D in vitro culture models for breast cancer research, specifically for adherence-based scaffolds (i.e. matrix-assisted). Finally, the future perspectives of 3D in vitro breast cancer models and their potential to lead to an improved understanding of breast cancer diagnosis and treatment are discussed.
Collapse
Affiliation(s)
- John Redmond
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland
| | - Helen McCarthy
- School of Pharmacy, Queen's University, Belfast BT9 7BL, United Kingdom; School of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| | - Paul Buchanan
- School of Nursing and Human Science, Dublin City University, Dublin 9, Ireland; National Institute of Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Tanya J Levingstone
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
44
|
Sahvieh S, Oryan A, Hassanajili S, Kamali A. Role of bone 1stem cell-seeded 3D polylactic acid/polycaprolactone/hydroxyapatite scaffold on a critical-sized radial bone defect in rat. Cell Tissue Res 2021; 383:735-750. [PMID: 32924069 DOI: 10.1007/s00441-020-03284-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/14/2020] [Indexed: 01/07/2023]
Abstract
Osteoconductive biomaterials were used to find the most reliable materials in bone healing. Our focus was on the bone healing capacity of the stem cell-loaded and unloaded PLA/PCL/HA scaffolds. The 3D scaffold of PLA/PCL/HA was characterized by scanning electron microscopy (SEM), rheology, X-ray diffraction (XRD), and Fourier transform-infrared (FT-IR) spectroscopy. Bone marrow stem cells (BMSCs) have multipotential differentiation into osteoblasts. Forty Wistar male rats were used to organize four experimental groups: control, autograft, scaffold, and BMSCs-loaded scaffold groups. qRT-PCR showed that the BMSCs-loaded scaffold had a higher expression level of CD31 and osteogenic markers compared with the control group (P < 0.05). Radiology and computed tomography (CT) scan evaluations showed significant improvement in the BMSCs-loaded scaffold compared with the control group (P < 0.001). Biomechanical estimation demonstrated significantly higher stress (P < 0.01), stiffness (P < 0.001), and ultimate load (P < 0.01) in the autograft and BMSCs-loaded scaffold groups compared with the untreated group and higher strain was seen in the control group than the other groups (P < 0.01). Histomorphometric and immunohistochemical (IHC) investigations showed significantly improved regeneration scores in the autograft and BMSCs-loaded scaffold groups compared with the control group (P < 0.05). Also, there was a significant difference between the scaffold and control groups in all tests (P < 0.05). The results depicted that our novel approach will allow to develop PLA/PCL/HA 3D scaffold in bone healing via BMSC loading.
Collapse
Affiliation(s)
- Sonia Sahvieh
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ahmad Oryan
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Shadi Hassanajili
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz, Iran
| | - Amir Kamali
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
45
|
Krüger-Genge A, Hauser S, Neffe AT, Liu Y, Lendlein A, Pietzsch J, Jung F. Response of Endothelial Cells to Gelatin-Based Hydrogels. ACS Biomater Sci Eng 2021; 7:527-540. [PMID: 33496571 DOI: 10.1021/acsbiomaterials.0c01432] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The establishment of confluent endothelial cell (EC) monolayers on implanted materials has been identified as a concept to avoid thrombus formation but is a continuous challenge in cardiovascular device engineering. Here, material properties of gelatin-based hydrogels obtained by reacting gelatin with varying amounts of lysine diisocyanate ethyl ester were correlated with the functional state of hydrogel contacting venous EC (HUVEC) and HUVEC's ability to form a monolayer on these hydrogels. The density of adherent HUVEC on the softest hydrogel at 37 °C (G' = 1.02 kPa, E = 1.1 ± 0.3 kPa) was significantly lower (125 mm-1) than on the stiffer hydrogels (920 mm-1; G' = 2.515 and 5.02 kPa, E = 4.8 ± 0.8 and 10.3 ± 1.2 kPa). This was accompanied by increased matrix metalloprotease activity (9 pmol·min-2 compared to 0.6 pmol·min-2) and stress fiber formation, while cell-to-cell contacts were comparable. Likewise, release of eicosanoids (e.g., prostacyclin release of 1.7 vs 0.2 pg·mL-1·cell-1) and the pro-inflammatory cytokine MCP-1 (8 vs <1.5 pg·mL-1·cell-1) was higher on the softer than on the stiffer hydrogels. The expressions of pro-inflammatory markers COX-2, COX-1, and RAGE were slightly increased on all hydrogels on day 2 (up to 200% of the control), indicating a weak inflammation; however, the levels dropped to below the control from day 6. The study revealed that hydrogels with higher moduli approached the status of a functionally confluent HUVEC monolayer. The results indicate the promising potential especially of the discussed gelatin-based hydrogels with higher G' as biomaterials for implants foreseen for the venous system.
Collapse
Affiliation(s)
- Anne Krüger-Genge
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328 Dresden, Germany
| | - Axel T Neffe
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Kantstr. 55, 14513 Teltow, Germany
| | - Yue Liu
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany
| | - Andreas Lendlein
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Kantstr. 55, 14513 Teltow, Germany.,Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328 Dresden, Germany.,School of Science, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01062 Dresden, Germany
| | - Friedrich Jung
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Helmholtz-Zentrum Geesthacht, Kantstr. 55, 14513 Teltow, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Kantstr. 55, 14513 Teltow, Germany
| |
Collapse
|
46
|
Schädli GN, Vetsch JR, Baumann RP, de Leeuw AM, Wehrle E, Rubert M, Müller R. Time-lapsed imaging of nanocomposite scaffolds reveals increased bone formation in dynamic compression bioreactors. Commun Biol 2021; 4:110. [PMID: 33495540 PMCID: PMC7835377 DOI: 10.1038/s42003-020-01635-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022] Open
Abstract
Progress in bone scaffold development relies on cost-intensive and hardly scalable animal studies. In contrast to in vivo, in vitro studies are often conducted in the absence of dynamic compression. Here, we present an in vitro dynamic compression bioreactor approach to monitor bone formation in scaffolds under cyclic loading. A biopolymer was processed into mechanically competent bone scaffolds that incorporate a high-volume content of ultrasonically treated hydroxyapatite or a mixture with barium titanate nanoparticles. After seeding with human bone marrow stromal cells, time-lapsed imaging of scaffolds in bioreactors revealed increased bone formation in hydroxyapatite scaffolds under cyclic loading. This stimulatory effect was even more pronounced in scaffolds containing a mixture of barium titanate and hydroxyapatite and corroborated by immunohistological staining. Therefore, by combining mechanical loading and time-lapsed imaging, this in vitro bioreactor strategy may potentially accelerate development of engineered bone scaffolds and reduce the use of animals for experimentation. Schädli et al. present a bioreactor system that combines mechanical loading with longitudinal microCT imaging to assess bone mineralization in a poly(lactic-co-glycolic acid) (PLGA) scaffold reinforced with nanoparticles. This approach allows rapid and rigorous evaluation of engineered bone scaffolds performance in vitro and might reduce the use of animals for experimentation.
Collapse
Affiliation(s)
- Gian Nutal Schädli
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Jolanda R Vetsch
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Robert P Baumann
- Particle Technology Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Anke M de Leeuw
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Esther Wehrle
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Marina Rubert
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
47
|
Entezari A, Swain MV, Gooding JJ, Roohani I, Li Q. A modular design strategy to integrate mechanotransduction concepts in scaffold-based bone tissue engineering. Acta Biomater 2020; 118:100-112. [PMID: 33059100 DOI: 10.1016/j.actbio.2020.10.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/01/2020] [Accepted: 10/08/2020] [Indexed: 12/19/2022]
Abstract
Repair or regeneration of load-bearing bones has long been an incentive for the tissue engineering community to develop a plethora of synthetic bone scaffolds. Despite the key role of physical forces and the mechanical environment in bone regeneration, the mechanotransduction concept has rarely been incorporated in structural design of bone tissue scaffolds, particularly those made of bioactive materials such as hydrogels and bioceramics. Herein, we introduce a modular design strategy to fabricate a load bearing device that can support a wide range of hydrogel- and ceramic-based scaffolds against complex in-vivo loading conditions to induce desirable mechanical strains for bone regeneration within the scaffolds. The device is comprised of a fenestrated polymeric shell and ceramic structural pillars arranged in a sophisticated configuration to provide ample internal space for the scaffold, also enabling it to purposely regulate the levels of strains and stresses within the scaffolds. Utilizing this top-down design approach, we demonstrate that the failure load of alginate hydrogels increases 3200-fold in compression, 300-fold in shear and 75-fold in impact, achieving the values that enable them to withstand physiological loads in weight-bearing sites, while allowing generation of osteoinductive strains (i.e., 0.2-0.4%) in the hydrogel. This modular design approach opens a broad range of opportunities to utilize various bioactive but mechanically weak scaffolds for the treatment of load-bearing defects and exploiting mechanobiology strategies to improve bone regeneration.
Collapse
|
48
|
Kampleitner C, Changi K, Felfel RM, Scotchford CA, Sottile V, Kluger R, Hoffmann O, Grant DM, Epstein MM. Preclinical biological and physicochemical evaluation of two-photon engineered 3D biomimetic copolymer scaffolds for bone healing. Biomater Sci 2020; 8:1683-1694. [PMID: 31984995 DOI: 10.1039/c9bm01827a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A major challenge in orthopedics is the repair of large non-union bone fractures. A promising therapy for this indication is the use of biodegradable bioinspired biomaterials that stabilize the fracture site, relieve pain and initiate bone formation and healing. This study uses a multidisciplinary evaluation strategy to assess immunogenicity, allergenicity, bone responses and physicochemical properties of a novel biomaterial scaffold. Two-photon stereolithography generated personalized custom-built scaffolds with a repeating 3D structure of Schwarz Primitive minimal surface unit cell with a specific pore size of ∼400 μm from three different methacrylated poly(d,l-lactide-co-ε-caprolactone) copolymers with lactide to caprolactone monomer ratios of 16 : 4, 18 : 2 and 9 : 1. Using in vitro and in vivo assays for bone responses, immunological reactions and degradation dynamics, we found that copolymer composition influenced the scaffold physicochemical and biological properties. The scaffolds with the fastest degradation rate correlated with adverse cellular effects and mechanical stiffness correlated with in vitro osteoblast mineralization. The physicochemical properties also correlated with in vivo bone healing and immune responses. Overall these observations provide compelling support for these scaffolds for bone repair and illustrate the effectiveness of a promising multidisciplinary strategy with great potential for the preclinical evaluation of biomaterials.
Collapse
Affiliation(s)
- Carina Kampleitner
- Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Katayoon Changi
- Laboratory of Experimental Allergy, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Department of Dermatology, Vienna, Austria.
| | - Reda M Felfel
- Advanced Materials Research Group, Faculty of Engineering, University of Nottingham, UK
| | - Colin A Scotchford
- Advanced Materials Research Group, Faculty of Engineering, University of Nottingham, UK
| | | | - Rainer Kluger
- Wolfson STEM Centre, School of Medicine, University of Nottingham, UK
| | - Oskar Hoffmann
- Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - David M Grant
- Advanced Materials Research Group, Faculty of Engineering, University of Nottingham, UK
| | - Michelle M Epstein
- Laboratory of Experimental Allergy, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Department of Dermatology, Vienna, Austria.
| |
Collapse
|
49
|
Liu E, Zhu D, Gonzalez Diaz E, Tong X, Yang F. Gradient Hydrogels for Optimizing Niche Cues to Enhance Cell-Based Cartilage Regeneration. Tissue Eng Part A 2020; 27:929-939. [PMID: 32940136 DOI: 10.1089/ten.tea.2020.0158] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Hydrogels have been widely used for cell delivery to enhance cell-based therapies for cartilage tissue regeneration. To better support cartilage deposition, it is imperative to determine hydrogel formulation with physical and biochemical cues that are optimized for different cell populations. Previous attempts to identify optimized hydrogels rely mostly on testing hydrogel formulations with discrete properties, which are time-consuming and require large amounts of cells and materials. Gradient hydrogels encompass a range of continuous changes in niche properties, therefore offering a promising solution for screening a wide range of cell-niche interactions using less materials and time. However, harnessing gradient hydrogels to assess how matrix stiffness modulates cartilage formation by different cell types in vivo have never been investigated before. The goal of this study is to fabricate gradient hydrogels for screening the effects of varying hydrogel stiffness on cartilage formation by mesenchymal stem cells (MSCs) and chondrocytes, respectively, the two most commonly used cell populations for cartilage regeneration. We fabricated stiffness gradient hydrogels with tunable dimensions that support homogeneous cell encapsulation. Using gradient hydrogels with tunable stiffness range, we found MSCs and chondrocytes exhibit opposite trend in cartilage deposition in response to stiffness changes in vitro. Specifically, MSCs require soft hydrogels with Young's modulus less than 5 kPa to support faster cartilage deposition, as shown by type II collagen and sulfated glycosaminoglycan staining. In contrast, chondrocytes produce cartilage more effectively in stiffer matrix (>20 kPa). We chose optimal ranges of stiffness for each cell population for further testing in vivo using a mouse subcutaneous model. Our results further validated that soft matrix (Young's modulus <5 kPa) is better in supporting MSC-based cartilage deposition in three-dimensional, whereas stiffer matrix (Young's modulus >20 kPa) is more desirable for supporting chondrocyte-based cartilage deposition. Our results show the importance of optimizing niche cues in a cell-type-specific manner and validate the potential of using gradient hydrogels for optimizing niche cues to support cartilage regeneration in vitro and in vivo. Impact statement The present study validates the utility of gradient hydrogels for determining optimal hydrogel stiffness for supporting cartilage regeneration using both chondrocytes and stem cells. We demonstrate that such gradient hydrogels can be used for fast optimizing matrix stiffness for specific cell type to support optimal cartilage regeneration. To our knowledge, this is the first demonstration of applying gradient hydrogels for assessing optimal niche cues that support tissue regeneration in vivo and may be used for assessing optimal niche cues for different cell types to regeneration of different tissues.
Collapse
Affiliation(s)
- Elisa Liu
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Danqing Zhu
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Eva Gonzalez Diaz
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Xinming Tong
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, California, USA.,Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| |
Collapse
|
50
|
Zhang J, Wehrle E, Adamek P, Paul GR, Qin XH, Rubert M, Müller R. Optimization of mechanical stiffness and cell density of 3D bioprinted cell-laden scaffolds improves extracellular matrix mineralization and cellular organization for bone tissue engineering. Acta Biomater 2020; 114:307-322. [PMID: 32673752 DOI: 10.1016/j.actbio.2020.07.016] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/02/2020] [Accepted: 07/08/2020] [Indexed: 12/22/2022]
Abstract
Bioprinting is an emerging technology in which cell-laden biomaterials are precisely dispersed to engineer artificial tissues that mimic aspects of the anatomical and structural complexity of relatively soft tissues such as skin, vessels, and cartilage. However, reproducing the highly mineralized and cellular diversity of bone tissue is still not easily achievable and is yet to be demonstrated. Here, an extrusion-based 3D bioprinting strategy is utilized to fabricate 3D bone-like tissue constructs containing osteogenic cellular organization. A simple and low-cost bioink for 3D bioprinting of bone-like tissue is prepared based on two unmodified polymers (alginate and gelatin) and combined with human mesenchymal stem cells (hMSCs). To form 3D bone-like tissue and bone cell phenotype, the influence of different scaffold stiffness and cell density of 3D bioprinted cell-laden porous scaffolds on osteogenic differentiation and bone-like tissue formation was investigated over time. Our results showed that soft scaffolds (0.8%alg, 0.66 ± 0.08 kPa) had higher DNA content, enhanced ALP activity and stimulated osteogenic differentiation than stiff scaffolds (1.8%alg, 5.4 ± 1.2 kPa). At day 42, significantly more mineralized tissue was formed in soft scaffolds than in stiff scaffolds (43.5 ± 7.1 mm3 vs. 22.6 ± 6.0 mm3). Importantly, immunohistochemistry staining demonstrated more osteocalcin protein expression in high mineral compared to low mineral regions. Additionally, cells in soft scaffolds exhibited osteoblast- and early osteocyte-related gene expression and 3D cellular network within the mineralized matrix at day 42. Furthermore, the results showed that cell density in 15 M cells/ml can promote cell-cell connections at day 7 and mineral formation at day 14, while 5 M cells/ml had the significantly higher mineral formation rate than 15 M cells/ml from day 14 to day 21. In summary, this work reports the formation of 3D bioprinted bone-like tissue using a simple and low-cost cell-laden bioink, which was optimized for stiffness and cell density, showing great promise for bone tissue engineering applications. STATEMENT OF SIGNIFICANCE: In this study, we presented for the first time a framework combining 3D bioprinting, bioreactor system and time-lapsed micro-CT monitoring to provide in vitro scaffold fabrication, maturation, and mineral visualization for bone tissue engineering. 3D bone-like tissue constructs have been formed via optimizing scaffold stiffness and cell density. The soft scaffolds had higher cell proliferation, enhanced alkaline phosphatase activity and stimulated osteogenic differentiation with 3D cellular network foramtion than stiff scaffolds. Significantly more mineralized bone-like tissue was formed in soft scaffolds than stiff scaffolds at day 42. Meanwhile, cell density in 15 M cells/ml can promote cell-cell connections and mineral formation in 14 days, while the higher mineral formation rate was found in 5 M cells/ml from day 14 to day 21.
Collapse
Affiliation(s)
- Jianhua Zhang
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093 Zurich, Switzerland
| | - Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093 Zurich, Switzerland
| | - Pavel Adamek
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093 Zurich, Switzerland
| | - Graeme R Paul
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093 Zurich, Switzerland
| | - Xiao-Hua Qin
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093 Zurich, Switzerland
| | - Marina Rubert
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093 Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093 Zurich, Switzerland.
| |
Collapse
|