1
|
Gao Q, Cekuc MS, Ergul YS, Pius AK, Shinohara I, Murayama M, Susuki Y, Ma C, Morita M, Chow SKH, Goodman SB. 3D Culture of MSCs for Clinical Application. Bioengineering (Basel) 2024; 11:1199. [PMID: 39768017 PMCID: PMC11726872 DOI: 10.3390/bioengineering11121199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 01/16/2025] Open
Abstract
Mesenchymal stem cells (MSCs) play an important role in regenerative medicine and drug discovery due to their multipotential differentiation capabilities and immunomodulatory effects. Compared with traditional 2D cultures of MSCs, 3D cultures of MSCs have emerged as an effective approach to enhance cell viability, proliferation, and functionality, and provide a more relevant physiological environment. Here, we review the therapeutic potential of 3D-cultured MSCs, highlighting their roles in tissue regeneration and repair and drug screening. We further summarize successful cases that apply 3D MSCs in modeling disease states, enabling the identification of novel therapeutic strategies. Despite these promising applications, we discuss challenges that remain in the clinical translation of 3D MSC technologies, including stability, cell heterogeneity, and regulatory issues. We conclude by addressing these obstacles and emphasizing the need for further research to fully exploit the potential of 3D MSCs in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94304, USA; (Q.G.)
| |
Collapse
|
2
|
Zhang H, Zhou Z, Zhang F, Wan C. Hydrogel-Based 3D Bioprinting Technology for Articular Cartilage Regenerative Engineering. Gels 2024; 10:430. [PMID: 39057453 PMCID: PMC11276275 DOI: 10.3390/gels10070430] [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: 05/13/2024] [Revised: 06/09/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Articular cartilage is an avascular tissue with very limited capacity of self-regeneration. Trauma or injury-related defects, inflammation, or aging in articular cartilage can induce progressive degenerative joint diseases such as osteoarthritis. There are significant clinical demands for the development of effective therapeutic approaches to promote articular cartilage repair or regeneration. The current treatment modalities used for the repair of cartilage lesions mainly include cell-based therapy, small molecules, surgical approaches, and tissue engineering. However, these approaches remain unsatisfactory. With the advent of three-dimensional (3D) bioprinting technology, tissue engineering provides an opportunity to repair articular cartilage defects or degeneration through the construction of organized, living structures composed of biomaterials, chondrogenic cells, and bioactive factors. The bioprinted cartilage-like structures can mimic native articular cartilage, as opposed to traditional approaches, by allowing excellent control of chondrogenic cell distribution and the modulation of biomechanical and biochemical properties with high precision. This review focuses on various hydrogels, including natural and synthetic hydrogels, and their current developments as bioinks in 3D bioprinting for cartilage tissue engineering. In addition, the challenges and prospects of these hydrogels in cartilage tissue engineering applications are also discussed.
Collapse
Affiliation(s)
- Hongji Zhang
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (H.Z.); (Z.Z.); (F.Z.)
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Key Laboratory of Regenerative Medicine (Shenzhen Base), Ministry of Education, School of Biomedical Sciences Core Laboratory, Institute of Stem Cell, Genomics and Translational Research, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518057, China
| | - Zheyuan Zhou
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (H.Z.); (Z.Z.); (F.Z.)
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Key Laboratory of Regenerative Medicine (Shenzhen Base), Ministry of Education, School of Biomedical Sciences Core Laboratory, Institute of Stem Cell, Genomics and Translational Research, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518057, China
| | - Fengjie Zhang
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (H.Z.); (Z.Z.); (F.Z.)
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Key Laboratory of Regenerative Medicine (Shenzhen Base), Ministry of Education, School of Biomedical Sciences Core Laboratory, Institute of Stem Cell, Genomics and Translational Research, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518057, China
| | - Chao Wan
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (H.Z.); (Z.Z.); (F.Z.)
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Key Laboratory of Regenerative Medicine (Shenzhen Base), Ministry of Education, School of Biomedical Sciences Core Laboratory, Institute of Stem Cell, Genomics and Translational Research, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
3
|
Zanrè E, Dalla Valle E, D’Angelo E, Sensi F, Agostini M, Cimetta E. Recent Advancements in Hydrogel Biomedical Research in Italy. Gels 2024; 10:248. [PMID: 38667667 PMCID: PMC11048829 DOI: 10.3390/gels10040248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/24/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Hydrogels have emerged as versatile biomaterials with remarkable applications in biomedicine and tissue engineering. Here, we present an overview of recent and ongoing research in Italy, focusing on extracellular matrix-derived, natural, and synthetic hydrogels specifically applied to biomedicine and tissue engineering. The analyzed studies highlight the versatile nature and wide range of applicability of hydrogel-based studies. Attention is also given to the integration of hydrogels within bioreactor systems, specialized devices used in biological studies to culture cells under controlled conditions, enhancing their potential for regenerative medicine, drug discovery, and drug delivery. Despite the abundance of literature on this subject, a comprehensive overview of Italian contributions to the field of hydrogels-based biomedical research is still missing and is thus our focus for this review. Consolidating a diverse range of studies, the Italian scientific community presents a complete landscape for hydrogel use, shaping the future directions of biomaterials research. This review aspires to serve as a guide and map for Italian researchers interested in the development and use of hydrogels in biomedicine.
Collapse
Affiliation(s)
- Eleonora Zanrè
- Department of Industrial Engineering (DII), University of Padova, 35131 Padova, Italy; (E.Z.); (E.D.V.)
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127 Padova, Italy; (E.D.); (F.S.); (M.A.)
| | - Eva Dalla Valle
- Department of Industrial Engineering (DII), University of Padova, 35131 Padova, Italy; (E.Z.); (E.D.V.)
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127 Padova, Italy; (E.D.); (F.S.); (M.A.)
| | - Edoardo D’Angelo
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127 Padova, Italy; (E.D.); (F.S.); (M.A.)
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
| | - Francesca Sensi
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127 Padova, Italy; (E.D.); (F.S.); (M.A.)
| | - Marco Agostini
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127 Padova, Italy; (E.D.); (F.S.); (M.A.)
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
| | - Elisa Cimetta
- Department of Industrial Engineering (DII), University of Padova, 35131 Padova, Italy; (E.Z.); (E.D.V.)
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127 Padova, Italy; (E.D.); (F.S.); (M.A.)
| |
Collapse
|
4
|
Shou Y, Teo XY, Wu KZ, Bai B, Kumar ARK, Low J, Le Z, Tay A. Dynamic Stimulations with Bioengineered Extracellular Matrix-Mimicking Hydrogels for Mechano Cell Reprogramming and Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300670. [PMID: 37119518 PMCID: PMC10375194 DOI: 10.1002/advs.202300670] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/10/2023] [Indexed: 06/19/2023]
Abstract
Cells interact with their surrounding environment through a combination of static and dynamic mechanical signals that vary over stimulus types, intensity, space, and time. Compared to static mechanical signals such as stiffness, porosity, and topography, the current understanding on the effects of dynamic mechanical stimulations on cells remains limited, attributing to a lack of access to devices, the complexity of experimental set-up, and data interpretation. Yet, in the pursuit of emerging translational applications (e.g., cell manufacturing for clinical treatment), it is crucial to understand how cells respond to a variety of dynamic forces that are omnipresent in vivo so that they can be exploited to enhance manufacturing and therapeutic outcomes. With a rising appreciation of the extracellular matrix (ECM) as a key regulator of biofunctions, researchers have bioengineered a suite of ECM-mimicking hydrogels, which can be fine-tuned with spatiotemporal mechanical cues to model complex static and dynamic mechanical profiles. This review first discusses how mechanical stimuli may impact different cellular components and the various mechanobiology pathways involved. Then, how hydrogels can be designed to incorporate static and dynamic mechanical parameters to influence cell behaviors are described. The Scopus database is also used to analyze the relative strength in evidence, ranging from strong to weak, based on number of published literatures, associated citations, and treatment significance. Additionally, the impacts of static and dynamic mechanical stimulations on clinically relevant cell types including mesenchymal stem cells, fibroblasts, and immune cells, are evaluated. The aim is to draw attention to the paucity of studies on the effects of dynamic mechanical stimuli on cells, as well as to highlight the potential of using a cocktail of various types and intensities of mechanical stimulations to influence cell fates (similar to the concept of biochemical cocktail to direct cell fate). It is envisioned that this progress report will inspire more exciting translational development of mechanoresponsive hydrogels for biomedical applications.
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
| | - Xin Yong Teo
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Kenny Zhuoran Wu
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Bingyu Bai
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Arun R. K. Kumar
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore117597Singapore
| | - Jessalyn Low
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Zhicheng Le
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
| | - Andy Tay
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
- NUS Tissue Engineering ProgramNational University of SingaporeSingapore117510Singapore
| |
Collapse
|
5
|
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: 0] [Impact Index Per Article: 0] [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
|
6
|
Vaca-González JJ, Culma JJS, Nova LMH, Garzón-Alvarado DA. Anatomy, molecular structures, and hyaluronic acid - Gelatin injectable hydrogels as a therapeutic alternative for hyaline cartilage recovery: A review. J Biomed Mater Res B Appl Biomater 2023. [PMID: 37178328 DOI: 10.1002/jbm.b.35261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/24/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
Cartilage damage caused by trauma or osteoarthritis is a common joint disease that can increase the social and economic burden in society. Due to its avascular characteristics, the poor migration ability of chondrocytes, and a low number of progenitor cells, the self-healing ability of cartilage defects has been significantly limited. Hydrogels have been developed into one of the most suitable biomaterials for the regeneration of cartilage because of its characteristics such as high-water absorption, biodegradation, porosity, and biocompatibility similar to natural extracellular matrix. Therefore, the present review article presents a conceptual framework that summarizes the anatomical, molecular structure and biochemical properties of hyaline cartilage located in long bones: articular cartilage and growth plate. Moreover, the importance of preparation and application of hyaluronic acid - gelatin hydrogels for cartilage tissue engineering are included. Hydrogels possess benefits of stimulating the production of Agc1, Col2α1-IIa, and SOX9, molecules important for the synthesis and composition of the extracellular matrix of cartilage. Accordingly, they are believed to be promising biomaterials of therapeutic alternatives to treat cartilage damage.
Collapse
Affiliation(s)
- Juan Jairo Vaca-González
- Escuela de Pregrado, Dirección Académica, Vicerrectoría de Sede, Universidad Nacional de Colombia, Sede de La Paz, Cesar, Colombia
- Biomimetics Laboratory, Biotechnology Institute, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Juan José Saiz Culma
- Biomimetics Laboratory, Biotechnology Institute, Universidad Nacional de Colombia, Bogotá, Colombia
| | | | - Diego Alexander Garzón-Alvarado
- Biomimetics Laboratory, Biotechnology Institute, Universidad Nacional de Colombia, Bogotá, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
7
|
Taheri S, Ghazali HS, Ghazali ZS, Bhattacharyya A, Noh I. Progress in biomechanical stimuli on the cell-encapsulated hydrogels for cartilage tissue regeneration. Biomater Res 2023; 27:22. [PMID: 36935512 PMCID: PMC10026525 DOI: 10.1186/s40824-023-00358-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 02/25/2023] [Indexed: 03/21/2023] Open
Abstract
BACKGROUND Worldwide, many people suffer from knee injuries and articular cartilage damage every year, which causes pain and reduces productivity, life quality, and daily routines. Medication is currently primarily used to relieve symptoms and not to ameliorate cartilage degeneration. As the natural healing capacity of cartilage damage is limited due to a lack of vascularization, common surgical methods are used to repair cartilage tissue, but they cannot prevent massive damage followed by injury. MAIN BODY Functional tissue engineering has recently attracted attention for the repair of cartilage damage using a combination of cells, scaffolds (constructs), biochemical factors, and biomechanical stimuli. As cyclic biomechanical loading is the key factor in maintaining the chondrocyte phenotype, many studies have evaluated the effect of biomechanical stimulation on chondrogenesis. The characteristics of hydrogels, such as their mechanical properties, water content, and cell encapsulation, make them ideal for tissue-engineered scaffolds. Induced cell signaling (biochemical and biomechanical factors) and encapsulation of cells in hydrogels as a construct are discussed for biomechanical stimulation-based tissue regeneration, and several notable studies on the effect of biomechanical stimulation on encapsulated cells within hydrogels are discussed for cartilage regeneration. CONCLUSION Induction of biochemical and biomechanical signaling on the encapsulated cells in hydrogels are important factors for biomechanical stimulation-based cartilage regeneration.
Collapse
Affiliation(s)
- Shiva Taheri
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea
| | - Hanieh Sadat Ghazali
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology, Tehran, 1684613114, Iran
| | - Zahra Sadat Ghazali
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, 158754413, Iran
| | - Amitava Bhattacharyya
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea
- Functional, Innovative, and Smart Textiles, PSG Institute of Advanced Studies, Coimbatore, 641004, India
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea
| | - Insup Noh
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea.
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul, 01811, Republic of Korea.
| |
Collapse
|
8
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
9
|
Xu W, Zhu J, Hu J, Xiao L. Engineering the biomechanical microenvironment of chondrocytes towards articular cartilage tissue engineering. Life Sci 2022; 309:121043. [DOI: 10.1016/j.lfs.2022.121043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/24/2022] [Accepted: 10/02/2022] [Indexed: 11/28/2022]
|
10
|
Ju DB, Lee JC, Hwang SK, Cho CS, Kim HJ. Progress of Polysaccharide-Contained Polyurethanes for Biomedical Applications. Tissue Eng Regen Med 2022; 19:891-912. [PMID: 35819712 PMCID: PMC9478012 DOI: 10.1007/s13770-022-00464-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/10/2022] [Accepted: 05/01/2022] [Indexed: 11/26/2022] Open
Abstract
Polyurethane (PU) has been widely examined and used for biomedical applications, such as catheters, blood oxygenators, stents, cardiac valves, drug delivery carriers, dialysis devices, wound dressings, adhesives, pacemaker, tissue engineering, and coatings for breast implants due to its mechanical flexibility, high tear strength, biocompatibility, and tailorable foams although bio-acceptability, biodegradability and controlled drug delivery to achieve the desired properties should be considered. Especially, during the last decade, the development of bio-based PUs has raised public awareness because of the concern with global plastic waste for creating more environmentally friended materials. Therefore, it is desirable to discuss polysaccharide (PS)-contained PU for the wound dressing and bone tissue engineering among bio-based PUs because PS has several advantages, such as biocompatibility, reproducibility from the natural resources, degradability, ease of incorporation of bioactive agents, ease of availability and cost-effectiveness, and structural feature of chemical modification to meet the desired needs to overcome the disadvantages of PU itself by containing the PS into the PU.
Collapse
Affiliation(s)
- Do-Bin Ju
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08824, Korea
| | - Jeong-Cheol Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08824, Korea
| | - Soo-Kyung Hwang
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08824, Korea
- Program in Environmental Materials Science, Department of Agriculture, Forestry and Bioresources, Seoul National University, Seoul, 08824, Korea
| | - Chong-Su Cho
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08824, Korea.
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08824, Korea.
| | - Hyun-Joong Kim
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08824, Korea.
- Program in Environmental Materials Science, Department of Agriculture, Forestry and Bioresources, Seoul National University, Seoul, 08824, Korea.
| |
Collapse
|
11
|
Bednarczyk E. Chondrocytes In Vitro Systems Allowing Study of OA. Int J Mol Sci 2022; 23:ijms231810308. [PMID: 36142224 PMCID: PMC9499487 DOI: 10.3390/ijms231810308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/17/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoarthritis (OA) is an extremely complex disease, as it combines both biological-chemical and mechanical aspects, and it also involves the entire joint consisting of various types of tissues, including cartilage and bone. This paper describes the methods of conducting cell cultures aimed at searching for the mechanical causes of OA development, therapeutic solutions, and methods of preventing the disease. It presents the systems for the cultivation of cartilage cells depending on the level of their structural complexity, and taking into account the most common solutions aimed at recreating the most important factors contributing to the development of OA, that is mechanical loads. In-vitro systems used in tissue engineering to investigate the phenomena associated with OA were specified depending on the complexity and purposefulness of conducting cell cultures.
Collapse
Affiliation(s)
- Ewa Bednarczyk
- Faculty of Mechanical and Industrial Engineering, Warsaw University of Technology, Narbutta 85, 02-524 Warsaw, Poland
| |
Collapse
|
12
|
Xu C, Hong Y. Rational design of biodegradable thermoplastic polyurethanes for tissue repair. Bioact Mater 2022; 15:250-271. [PMID: 35386346 PMCID: PMC8940769 DOI: 10.1016/j.bioactmat.2021.11.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/09/2021] [Accepted: 11/24/2021] [Indexed: 12/25/2022] Open
Abstract
As a type of elastomeric polymers, non-degradable polyurethanes (PUs) have a long history of being used in clinics, whereas biodegradable PUs have been developed in recent decades, primarily for tissue repair and regeneration. Biodegradable thermoplastic (linear) PUs are soft and elastic polymeric biomaterials with high mechanical strength, which mimics the mechanical properties of soft and elastic tissues. Therefore, biodegradable thermoplastic polyurethanes are promising scaffolding materials for soft and elastic tissue repair and regeneration. Generally, PUs are synthesized by linking three types of changeable blocks: diisocyanates, diols, and chain extenders. Alternating the combination of these three blocks can finely tailor the physio-chemical properties and generate new functional PUs. These PUs have excellent processing flexibilities and can be fabricated into three-dimensional (3D) constructs using conventional and/or advanced technologies, which is a great advantage compared with cross-linked thermoset elastomers. Additionally, they can be combined with biomolecules to incorporate desired bioactivities to broaden their biomedical applications. In this review, we comprehensively summarized the synthesis, structures, and properties of biodegradable thermoplastic PUs, and introduced their multiple applications in tissue repair and regeneration. A whole picture of their design and applications along with discussions and perspectives of future directions would provide theoretical and technical supports to inspire new PU development and novel applications.
Collapse
Affiliation(s)
- Cancan Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| |
Collapse
|
13
|
Banh L, Cheung KK, Chan MWY, Young EWK, Viswanathan S. Advances in organ-on-a-chip systems for modelling joint tissue and osteoarthritic diseases. Osteoarthritis Cartilage 2022; 30:1050-1061. [PMID: 35460872 DOI: 10.1016/j.joca.2022.03.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
Joint-on-a-chip (JOC) models are powerful tools that aid in osteoarthritis (OA) research. These microfluidic devices apply emerging organ-on-a-chip technology to recapitulate a multifaceted joint tissue microenvironment. JOCs address the need for advanced, dynamic in vitro models that can mimic the in vivo tissue environment through joint-relevant biomechanical or fluidic integration, an aspect that existing in vitro OA models lack. There are existing review articles on OA models that focus on animal, tissue explant, and two-dimensional and three-dimensional (3D) culture systems, including microbioreactors and 3D printing technology, but there has been limited discussion of JOC models. The aim of this article is to review recent developments in human JOC technology and identify gaps for future advancements. Specifically, mechanical stimulation systems that mimic articular movement, multi-joint tissue cultures that enable crosstalk, and systems that aim to capture aspects of OA inflammation by incorporating immune cells are covered. The development of an advanced JOC model that captures the dynamic joint microenvironment will improve testing and translation of potential OA therapeutics.
Collapse
Affiliation(s)
- L Banh
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Canada; Krembil Research Institute, University Health Network, Canada; Institute of Biomedical Engineering, University of Toronto, Canada.
| | - K K Cheung
- Department of Mechanical & Industrial Engineering, University of Toronto, Canada.
| | - M W Y Chan
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Canada; Krembil Research Institute, University Health Network, Canada; Institute of Biomedical Engineering, University of Toronto, Canada.
| | - E W K Young
- Institute of Biomedical Engineering, University of Toronto, Canada; Department of Mechanical & Industrial Engineering, University of Toronto, Canada.
| | - S Viswanathan
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Canada; Krembil Research Institute, University Health Network, Canada; Institute of Biomedical Engineering, University of Toronto, Canada; Division of Hematology, Department of Medicine, University of Toronto, Canada.
| |
Collapse
|
14
|
Guimarães CF, Marques AP, Reis RL. Pushing the Natural Frontier: Progress on the Integration of Biomaterial Cues toward Combinatorial Biofabrication and Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105645. [PMID: 35419887 DOI: 10.1002/adma.202105645] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/14/2022] [Indexed: 06/14/2023]
Abstract
The engineering of fully functional, biological-like tissues requires biomaterials to direct cellular events to a near-native, 3D niche extent. Natural biomaterials are generally seen as a safe option for cell support, but their biocompatibility and biodegradability can be just as limited as their bioactive/biomimetic performance. Furthermore, integrating different biomaterial cues and their final impact on cellular behavior is a complex equation where the outcome might be very different from the sum of individual parts. This review critically analyses recent progress on biomaterial-induced cellular responses, from simple adhesion to more complex stem cell differentiation, looking at the ever-growing possibilities of natural materials modification. Starting with a discussion on native material formulation and the inclusion of cell-instructive cues, the roles of shape and mechanical stimuli, the susceptibility to cellular remodeling, and the often-overlooked impact of cellular density and cell-cell interactions within constructs, are delved into. Along the way, synergistic and antagonistic combinations reported in vitro and in vivo are singled out, identifying needs and current lessons on the development of natural biomaterial libraries to solve the cell-material puzzle efficiently. This review brings together knowledge from different fields envisioning next-generation, combinatorial biomaterial development toward complex tissue engineering.
Collapse
Affiliation(s)
- Carlos F Guimarães
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra P Marques
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
15
|
Bonifacio MA, Cometa S, Cochis A, Scalzone A, Gentile P, Scalia AC, Rimondini L, Mastrorilli P, De Giglio E. A bioprintable gellan gum/lignin hydrogel: a smart and sustainable route for cartilage regeneration. Int J Biol Macromol 2022; 216:336-346. [PMID: 35798077 DOI: 10.1016/j.ijbiomac.2022.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022]
Abstract
In this work a hydrogel, based on a blend of two gellan gums with different acyl content embedding lignin (up to 0.4%w/v) and crosslinked with magnesium ions, was developed for cartilage regeneration. The physico-chemical characterizations established that no chemical interaction between lignin and polysaccharides was detected. Lignin achieved up to 80 % of ascorbic acid's radical scavenging activity in vitro on DPPH and ABTS radicals. Viability of hMSC onto hydrogel containing lignin resulted comparable to the lignin-free one (>70 % viable cells, p > 0.05). The presence of lignin improved the hMSC 3D-constructs chondrogenesis, bringing to a significant (p < 0.05) up-regulation of the collagen type II, aggrecan and SOX 9 chondrogenic genes, and conferred bacteriostatic properties to the hydrogel, reducing the proliferation of S. aureus and S. epidermidis. Finally, cellularized 3D-constructs were manufactured via 3D-bioprinting confirming the processability of the formulation as a bioink and its unique biological features for creating a physiological milieu for cell growth.
Collapse
Affiliation(s)
- Maria A Bonifacio
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy; INSTM, National Consortium of Materials Science and Technology, Via G. Giusti 9, 50121 Florence, Italy.
| | | | - Andrea Cochis
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Health Sciences, 28100 Novara, Italy.
| | - Annachiara Scalzone
- Newcastle University, School of Engineering, Claremont Road, NE1 7RU Newcastle upon Tyne, United Kingdom.
| | - Piergiorgio Gentile
- Newcastle University, School of Engineering, Claremont Road, NE1 7RU Newcastle upon Tyne, United Kingdom.
| | - Alessandro C Scalia
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Health Sciences, 28100 Novara, Italy.
| | - Lia Rimondini
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Health Sciences, 28100 Novara, Italy.
| | - Piero Mastrorilli
- DICATECh Department Politecnico di Bari, Via Orabona 4, 70126 Bari, Italy.
| | - Elvira De Giglio
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy; INSTM, National Consortium of Materials Science and Technology, Via G. Giusti 9, 50121 Florence, Italy.
| |
Collapse
|
16
|
Volz M, Wyse-Sookoo KR, Travascio F, Huang CY, Best TM. MECHANOBIOLOGICAL APPROACHES FOR STIMULATING CHONDROGENESIS OF STEM CELLS. Stem Cells Dev 2022; 31:460-487. [PMID: 35615879 DOI: 10.1089/scd.2022.0049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Chondrogenesis is the process of differentiation of stem cells into mature chondrocytes. Such a process consists of chemical, functional, and structural changes which are initiated and mediated by the host environment of the cells. To date, the mechanobiology of chondrogenesis has not been fully elucidated. Hence, experimental activity is focused on recreating specific environmental conditions for stimulating chondrogenesis, and to look for a mechanistic interpretation of the mechanobiological response of cells in the cartilaginous tissues. There are a large number of studies on the topic that vary considerably in their experimental protocols used for providing environmental cues to cells for differentiation, making generalizable conclusions difficult to ascertain. The main objective of this contribution is to review the mechanobiological stimulation of stem cell chondrogenesis and methodological approaches utilized to date to promote chondrogenesis of stem cells in-vitro. In-vivo models will also be explored, but this area is currently limited. An overview of the experimental approaches used by different research groups may help the development of unified testing methods that could be used to overcome existing knowledge gaps, leading to an accelerated translation of experimental findings to clinical practice.
Collapse
Affiliation(s)
- Mallory Volz
- University of Miami, 5452, Biomedical Engineering, Coral Gables, Florida, United States;
| | | | - Francesco Travascio
- University of Miami, 5452, Mechanical and Aerospace Engineering, 1251 Memorial Drive, MEB 217B, Coral Gables, Florida, United States, 33146;
| | - Chun-Yuh Huang
- University of Miami, 5452, Biomedical Engineering, Coral Gables, Florida, United States;
| | - Thomas M Best
- University of Miami Miller School of Medicine, 12235, School of Medicine, Miami, Florida, United States;
| |
Collapse
|
17
|
Mackin RT, Fontenot KR, Edwards JV, Prevost NT, Jordan JH, Easson MW, Condon BD, French AD. Detection of Human Neutrophil Elastase by Fluorescent Peptide Sensors Conjugated to TEMPO-Oxidized Nanofibrillated Cellulose. Int J Mol Sci 2022; 23:3101. [PMID: 35328520 PMCID: PMC8952216 DOI: 10.3390/ijms23063101] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/22/2022] [Accepted: 03/02/2022] [Indexed: 12/21/2022] Open
Abstract
Peptide-cellulose conjugates designed for use as optical protease sensors have gained interest for point-of-care (POC) detection. Elevated serine protease levels are often found in patients with chronic illnesses, necessitating optimal biosensor design for POC assessment. Nanocellulose provides a platform for protease sensors as a transducer surface, and the employment of nanocellulose in this capacity combines its biocompatibility and high specific surface area properties to confer sensitive detection of dilute biomarkers. However, a basic understanding of the spatiotemporal relationships of the transducer surface and sensor disposition is needed to improve protease sensor design and development. Here, we examine a tripeptide, fluorogenic elastase biosensor attached to TEMPO-oxidized nanofibrillated cellulose via a polyethylene glycol linker. The synthetic conjugate was found to be active in the presence of human neutrophil elastase at levels comparable to other cellulose-based biosensors. Computational models examined the relationship of the sensor molecule to the transducer surface. The results illustrate differences in two crystallite transducer surfaces ((110) vs. (1-10)) and reveal preferred orientations of the sensor. Finally, a determination of the relative (110) vs. (1-10) orientations of crystals extracted from cotton demonstrates a preference for the (1-10) conformer. This model study potentiates the HNE sensor results for enhanced sensor activity design.
Collapse
Affiliation(s)
| | | | - Judson Vincent Edwards
- United States Department of Agriculture, Agriculture Research Service, Southern Regional Research Center (USDA-ARS-SRRC), New Orleans, LA 70124, USA; (R.T.M.); (K.R.F.); (N.T.P.); (J.H.J.); (M.W.E.); (B.D.C.); (A.D.F.)
| | | | | | | | | | | |
Collapse
|
18
|
Phelps J, Leonard C, Shah S, Krawetz R, Hart DA, Duncan NA, Sen A. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:73-87. [PMID: 35641171 PMCID: PMC8895489 DOI: 10.1093/stcltm/szab008] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal progenitor cells (MPCs) have shown promise initiating articular cartilage repair, with benefits largely attributed to the trophic factors they secrete. These factors can be found in the conditioned medium (CM) collected from cell cultures, and it is believed that extracellular vesicles (EVs) within this CM are at least partially responsible for MPC therapeutic efficacy. This study aimed to examine the functionality of the EV fraction of CM compared to whole CM obtained from human adipose-derived MPCs in an in vivo murine cartilage defect model. Mice treated with whole CM or the EV fraction demonstrated an enhanced cartilage repair score and type II collagen deposition at the injury site compared to saline controls. We then developed a scalable bioprocess using stirred suspension bioreactors (SSBs) to generate clinically relevant quantities of MPC-EVs. Whereas static monolayer culture systems are simple to use and readily accessible, SSBs offer increased scalability and a more homogenous environment due to constant mixing. This study evaluated the biochemical and functional properties of MPCs and their EV fractions generated in static culture versus SSBs. Functionality was assessed using in vitro MPC chondrogenesis as an outcome measure. SSBs supported increased MPC expression of cartilage-specific genes, and EV fractions derived from both static and SSB culture systems upregulated type II collagen production by MPCs. These results suggest that SSBs are an effective platform for the generation of MPC-derived EVs with the potential to induce cartilage repair.
Collapse
Affiliation(s)
- Jolene Phelps
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Catherine Leonard
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Sophia Shah
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Roman Krawetz
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - David A Hart
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Neil A Duncan
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Musculoskeletal Mechanobiology and Multiscale Mechanics Bioengineering Lab, Department of Civil Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Arindom Sen
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Center for Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Corresponding author: Arindom Sen, Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada. Tel: +403-210-9452; Fax: +403-220-8962;
| |
Collapse
|
19
|
Daou F, Cochis A, Leigheb M, Rimondini L. Current Advances in the Regeneration of Degenerated Articular Cartilage: A Literature Review on Tissue Engineering and Its Recent Clinical Translation. MATERIALS 2021; 15:ma15010031. [PMID: 35009175 PMCID: PMC8745794 DOI: 10.3390/ma15010031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 12/23/2022]
Abstract
Functional ability is the basis of healthy aging. Articular cartilage degeneration is amongst the most prevalent degenerative conditions that cause adverse impacts on the quality of life; moreover, it represents a key predisposing factor to osteoarthritis (OA). Both the poor capacity of articular cartilage for self-repair and the unsatisfactory outcomes of available clinical interventions make innovative tissue engineering a promising therapeutic strategy for articular cartilage repair. Significant progress was made in this field; however, a marked heterogeneity in the applied biomaterials, biofabrication, and assessments is nowadays evident by the huge number of research studies published to date. Accordingly, this literature review assimilates the most recent advances in cell-based and cell-free tissue engineering of articular cartilage and also focuses on the assessments performed via various in vitro studies, ex vivo models, preclinical in vivo animal models, and clinical studies in order to provide a broad overview of the latest findings and clinical translation in the context of degenerated articular cartilage and OA.
Collapse
Affiliation(s)
- Farah Daou
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (F.D.); (A.C.); (M.L.)
| | - Andrea Cochis
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (F.D.); (A.C.); (M.L.)
| | - Massimiliano Leigheb
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (F.D.); (A.C.); (M.L.)
- Department of Orthopaedics and Traumatology, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Lia Rimondini
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (F.D.); (A.C.); (M.L.)
- Correspondence: ; Tel.: +39-0321-660-673
| |
Collapse
|
20
|
Varshosaz J, Sajadi-Javan ZS, Kouhi M, Mirian M. Effect of bassorin (derived from gum tragacanth) and halloysite nanotubes on physicochemical properties and the osteoconductivity of methylcellulose-based injectable hydrogels. Int J Biol Macromol 2021; 192:869-882. [PMID: 34634330 DOI: 10.1016/j.ijbiomac.2021.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 09/24/2021] [Accepted: 10/03/2021] [Indexed: 12/15/2022]
Abstract
Injectable hydrogels have been known as promising materials for the regeneration of irregular shape tissue defects. In this study, novel thermosensitive methylcellulose (MC) hydrogels containing bassorin (Ba) and halloysite nanotubes (HNTs) have been developed for application in bone tissue engineering. Bassorin isolated from gum tragacanth (GT) with the concentration of 0.25-1.5 w/v% was blended with MC. The best MC/Ba gel (containing 0.5% bassorin) was chosen based on the results of injectability and rheological tests. HNTs (1-7%) were added to this formulation and tested for the physicochemical, mechanical, rheological, degradation, swelling, and biological properties. In vitro biological evaluations including cell proliferation (by MTT assay), cell attachment (by SEM), osteogenic activity (by Alizarin Red staining and alkaline phosphatase assay), and osteogenic gene expression (by quantitative real-time polymerase chain reaction) were done using MG-63 cells. Results showed that bassorin led to the increased gel-forming ability (at a lower temperature) and mechanical properties of MC hydrogel. The presence of HNTs and bassorin affected the degradation rate and swelling degree of MC-based hydrogel. Results showed significant enhancement in cell proliferation, differentiation, and mineralization, as well as higher bone-specific gene expression of the cell on bassorin and HNTs incorporated MC compared to pure MC hydrogel.
Collapse
Affiliation(s)
- Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Zahra Sadat Sajadi-Javan
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Monireh Kouhi
- Dental Materials Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
21
|
Shestovskaya MV, Bozhkova SA, Sopova JV, Khotin MG, Bozhokin MS. Methods of Modification of Mesenchymal Stem Cells and Conditions of Their Culturing for Hyaline Cartilage Tissue Engineering. Biomedicines 2021; 9:biomedicines9111666. [PMID: 34829895 PMCID: PMC8615732 DOI: 10.3390/biomedicines9111666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
The use of mesenchymal stromal cells (MSCs) for tissue engineering of hyaline cartilage is a topical area of regenerative medicine that has already entered clinical practice. The key stage of this procedure is to create conditions for chondrogenic differentiation of MSCs, increase the synthesis of hyaline cartilage extracellular matrix proteins by these cells and activate their proliferation. The first such works consisted in the indirect modification of cells, namely, in changing the conditions in which they are located, including microfracturing of the subchondral bone and the use of 3D biodegradable scaffolds. The most effective methods for modifying the cell culture of MSCs are protein and physical, which have already been partially introduced into clinical practice. Genetic methods for modifying MSCs, despite their effectiveness, have significant limitations. Techniques have not yet been developed that allow studying the effectiveness of their application even in limited groups of patients. The use of MSC modification methods allows precise regulation of cell culture proliferation, and in combination with the use of a 3D biodegradable scaffold, it allows obtaining a hyaline-like regenerate in the damaged area. This review is devoted to the consideration and comparison of various methods used to modify the cell culture of MSCs for their use in regenerative medicine of cartilage tissue.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Svetlana A. Bozhkova
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
| | - Julia V. Sopova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Mikhail G. Khotin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Mikhail S. Bozhokin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
22
|
Zhong Y, Caplan AI, Welter JF, Baskaran H. Glucose Availability Affects Extracellular Matrix Synthesis During Chondrogenesis In Vitro. Tissue Eng Part A 2021; 27:1321-1332. [PMID: 33499734 PMCID: PMC8610032 DOI: 10.1089/ten.tea.2020.0144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 01/22/2021] [Indexed: 01/09/2023] Open
Abstract
Understanding in vitro chondrogenesis of human mesenchymal stem cells (hMSCs) is important as it holds great promise for cartilage tissue engineering and other applications. The current technology produces the end tissue quality that is highly variable and dependent on culture conditions. We investigated the effect of nutrient availability on hMSC chondrogenesis in a static aggregate culture system by varying the medium-change frequency together with starting glucose levels. Glucose uptake and lactate secretion profiles were obtained to monitor the metabolism change during hMSC chondrogenesis with different culture conditions. Higher medium-change frequency led to increases in cumulative glucose uptake for all starting glucose levels. Furthermore, increase in glucose uptake by aggregates led to increased end tissue glycosaminoglycan (GAG) and hydroxyproline (HYP) content. The results suggest that increased glucose availability either through increased medium-change frequency or higher initial glucose levels lead to improved chondrogenesis. Also, cumulative glucose uptake and lactate secretion were found to correlate well with GAG and HYP content, indicating both molecules are promising biomarkers for noninvasive assessment of hMSC chondrogenesis. Collectively, our results can be used to design optimal culture conditions and develop dynamic assessment strategies for cartilage tissue engineering applications. Impact statement In this study, we investigated how culture conditions, medium-change frequency and glucose levels, affect chondrogenesis of human mesenchymal stem cells in an aggregate culture model. Doubling the medium-change frequency significantly increased the biochemical quality of the resultant tissue aggregates, as measured by their glycosaminoglycan and hydroxyproline content. We attribute this to increased glucose uptake through the glycolysis pathway, as secretion of lactate, a key endpoint product of the glycolysis pathway, increased concurrently. These findings can be used to design optimal culture conditions for tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- CWRU Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, Ohio, USA
| | - Arnold I. Caplan
- CWRU Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biology and Case Western Reserve University, Cleveland, Ohio, USA
| | - Jean F. Welter
- CWRU Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biology and Case Western Reserve University, Cleveland, Ohio, USA
| | - Harihara Baskaran
- CWRU Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
23
|
Casari G, Resca E, Giorgini A, Candini O, Petrachi T, Piccinno MS, Foppiani EM, Pacchioni L, Starnoni M, Pinelli M, De Santis G, Selleri F, Catani F, Dominici M, Veronesi E. Microfragmented adipose tissue is associated with improved ex vivo performance linked to HOXB7 and b-FGF expression. Stem Cell Res Ther 2021; 12:481. [PMID: 34454577 PMCID: PMC8399787 DOI: 10.1186/s13287-021-02540-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 08/02/2021] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Adipose tissue (AT) has become a source of mesenchymal stromal/stem cells (MSC) for regenerative medicine applications, in particular skeletal disorders. Several enzymatic or mechanical procedures have been proposed to process AT with the aim to isolate cells that can be locally implanted. How AT is processed may impact its properties. Thus, we compared AT processed by centrifugation (C-AT) to microfragmentation (MF-AT). Focusing on MF-AT, we subsequently assessed the impact of synovial fluid (SF) alone on both MF-AT and isolated AT-MSC to better understand their cartilage repair mechanisms. MATERIALS AND METHODS MF-AT and C-AT from the same donors were compared by histology and qRT-PCR immediately after isolation or as ex vivo cultures using a micro-tissue pellet system. The in vitro impact of SF on MF-AT and AT-MSC was assessed by histological staining and molecular analysis. RESULTS The main AT histological features (i.e., increased extracellular matrix and cellularity) of the freshly isolated or ex vivo-cultured MF-AT persisted compared to C-AT, which rapidly deteriorated during culture. Based on our previous studies of HOX genes in MSC, we investigated the involvement of Homeobox Protein HOX-B7 (HOXB7) and its target basic Fibroblast Growth Factor (bFGF) in the molecular mechanism underlying the improved performance of MF-AT. Indeed, both these biomarkers were more prominent in freshly isolated MF-AT compared to C-AT. SF alone preserved the AT histological features of MF-AT, together with HOXB7 and bFGF expression. Increased cell performance was also observed in isolated AT-MSC after SF treatment concomitant with enhanced HOXB7 expression, although there was no apparent association with bFGF. CONCLUSIONS Our findings show that MF has a positive effect on the maintenance of AT histology and may trigger the expression of trophic factors that improve tissue repair by processed AT.
Collapse
Affiliation(s)
- Giulia Casari
- Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy.,Rigenerand srl, Medolla, Modena, Italy
| | - Elisa Resca
- Technopole Mario Veronesi, Mirandola, Modena, Italy
| | - Andrea Giorgini
- Department of Orthopaedic and Traumatology, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | - Lucrezia Pacchioni
- Division of Plastic Surgery, Department of General Surgery and Surgical Specialties, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Marta Starnoni
- Division of Plastic Surgery, Department of General Surgery and Surgical Specialties, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Pinelli
- Division of Plastic Surgery, Department of General Surgery and Surgical Specialties, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Giorgio De Santis
- Division of Plastic Surgery, Department of General Surgery and Surgical Specialties, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Filippo Selleri
- Department of Orthopaedic and Traumatology, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Fabio Catani
- Department of Orthopaedic and Traumatology, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy. .,Rigenerand srl, Medolla, Modena, Italy. .,Technopole Mario Veronesi, Mirandola, Modena, Italy.
| | - Elena Veronesi
- Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy. .,Technopole Mario Veronesi, Mirandola, Modena, Italy.
| |
Collapse
|
24
|
Szustak M, Gendaszewska-Darmach E. Nanocellulose-Based Scaffolds for Chondrogenic Differentiation and Expansion. Front Bioeng Biotechnol 2021; 9:736213. [PMID: 34485266 PMCID: PMC8415884 DOI: 10.3389/fbioe.2021.736213] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Nanocellulose deserves special attention among the large group of biocompatible biomaterials. It exhibits good mechanical properties, which qualifies it for potential use as a scaffold imitating cartilage. However, the reconstruction of cartilage is a big challenge due to this tissue's limited regenerative capacity resulting from its lack of vascularization, innervations, and sparsely distributed chondrocytes. This feature restricts the infiltration of progenitor cells into damaged sites. Unfortunately, differentiated chondrocytes are challenging to obtain, and mesenchymal stem cells have become an alternative approach to promote chondrogenesis. Importantly, nanocellulose scaffolds induce the differentiation of stem cells into chondrocyte phenotypes. In this review, we present the recent progress of nanocellulose-based scaffolds promoting the development of cartilage tissue, especially within the emphasis on chondrogenic differentiation and expansion.
Collapse
Affiliation(s)
| | - Edyta Gendaszewska-Darmach
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Lodz, Poland
| |
Collapse
|
25
|
Jauković A, Abadjieva D, Trivanović D, Stoyanova E, Kostadinova M, Pashova S, Kestendjieva S, Kukolj T, Jeseta M, Kistanova E, Mourdjeva M. Specificity of 3D MSC Spheroids Microenvironment: Impact on MSC Behavior and Properties. Stem Cell Rev Rep 2021; 16:853-875. [PMID: 32681232 DOI: 10.1007/s12015-020-10006-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSC) have been considered the promising candidates for the regenerative and personalized medicine due to their self-renewal potential, multilineage differentiation and immunomodulatory capacity. Although these properties have encouraged profound MSC studies in recent years, the majority of research has been based on standard 2D culture utilization. The opportunity to resemble in vivo characteristics of cells native niche has been provided by implementation of 3D culturing models such as MSC spheroid formation assesed through cells self-assembling. In this review, we address the current literature on physical and biochemical features of 3D MSC spheroid microenvironment and their impact on MSC properties and behaviors. Starting with the reduction in the cells' dimensions and volume due to the changes in adhesion molecules expression and cytoskeletal proteins rearrangement resembling native conditions, through the microenvironment shifts in oxygen, nutrients and metabolites gradients and demands, we focus on distinctive and beneficial features of MSC in spheroids compared to cells cultured in 2D conditions. By summarizing the data for 3D MSC spheroids regarding cell survival, pluripotency, differentiation, immunomodulatory activities and potential to affect tumor cells growth we highlighted advantages and perspectives of MSC spheroids use in regenerative medicine. Further detailed analyses are needed to deepen our understanding of mechanisms responsible for modified MSC behavior in spheroids and to set future directions for MSC clinical application.
Collapse
Affiliation(s)
- Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, PO BOX 102, Belgrade, 11129, Serbia
| | - Desislava Abadjieva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Drenka Trivanović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, PO BOX 102, Belgrade, 11129, Serbia.,IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics, Röntgenring 11, D-97070, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Germany
| | - Elena Stoyanova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Milena Kostadinova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Shina Pashova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Snejana Kestendjieva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Tamara Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, PO BOX 102, Belgrade, 11129, Serbia
| | - Michal Jeseta
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Obilní trh 11, 602 00, Brno, Czech Republic.,Department of Veterinary Sciences, Czech University of Life Sciences in Prague, Kamýcká 129, 165 00, Suchdol, Praha 6, Czech Republic
| | - Elena Kistanova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria
| | - Milena Mourdjeva
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shoes, 1113, Sofia, Bulgaria.
| |
Collapse
|
26
|
Gao Y, Peng K, Mitragotri S. Covalently Crosslinked Hydrogels via Step-Growth Reactions: Crosslinking Chemistries, Polymers, and Clinical Impact. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2006362. [PMID: 33988273 DOI: 10.1002/adma.202006362] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/24/2020] [Indexed: 06/12/2023]
Abstract
Hydrogels are an important class of biomaterials with the unique property of high-water content in a crosslinked polymer network. In particular, chemically crosslinked hydrogels have made a great clinical impact in past years because of their desirable mechanical properties and tunability of structural and chemical properties. Various polymers and step-growth crosslinking chemistries are harnessed for fabricating such covalently crosslinked hydrogels for translational research. However, selecting appropriate crosslinking chemistries and polymers for the intended clinical application is time-consuming and challenging. It requires the integration of polymer chemistry knowledge with thoughtful crosslinking reaction design. This task becomes even more challenging when other factors such as the biological mechanisms of the pathology, practical administration routes, and regulatory requirements add additional constraints. In this review, key features of crosslinking chemistries and polymers commonly used for preparing translatable hydrogels are outlined and their performance in biological systems is summarized. The examples of effective polymer/crosslinking chemistry combinations that have yielded clinically approved hydrogel products are specifically highlighted. These hydrogel design parameters in the context of the regulatory process and clinical translation barriers, providing a guideline for the rational selection of polymer/crosslinking chemistry combinations to construct hydrogels with high translational potential are further considered.
Collapse
Affiliation(s)
- Yongsheng Gao
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute of Biologically Inspired Engineering, Boston, MA, 02115, USA
| | - Kevin Peng
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute of Biologically Inspired Engineering, Boston, MA, 02115, USA
| | - Samir Mitragotri
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute of Biologically Inspired Engineering, Boston, MA, 02115, USA
| |
Collapse
|
27
|
Arora S, Srinivasan A, Leung CM, Toh YC. Bio-mimicking Shear Stress Environments for Enhancing Mesenchymal Stem Cell Differentiation. Curr Stem Cell Res Ther 2021; 15:414-427. [PMID: 32268869 DOI: 10.2174/1574888x15666200408113630] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/03/2019] [Accepted: 02/19/2020] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells, with the ability to differentiate into mesodermal (e.g., adipocyte, chondrocyte, hematopoietic, myocyte, osteoblast), ectodermal (e.g., epithelial, neural) and endodermal (e.g., hepatocyte, islet cell) lineages based on the type of induction cues provided. As compared to embryonic stem cells, MSCs hold a multitude of advantages from a clinical translation perspective, including ease of isolation, low immunogenicity and limited ethical concerns. Therefore, MSCs are a promising stem cell source for different regenerative medicine applications. The in vitro differentiation of MSCs into different lineages relies on effective mimicking of the in vivo milieu, including both biochemical and mechanical stimuli. As compared to other biophysical cues, such as substrate stiffness and topography, the role of fluid shear stress (SS) in regulating MSC differentiation has been investigated to a lesser extent although the role of interstitial fluid and vascular flow in regulating the normal physiology of bone, muscle and cardiovascular tissues is well-known. This review aims to summarise the current state-of-the-art regarding the role of SS in the differentiation of MSCs into osteogenic, cardiovascular, chondrogenic, adipogenic and neurogenic lineages. We will also highlight and discuss the potential of employing SS to augment the differentiation of MSCs to other lineages, where SS is known to play a role physiologically but has not yet been successfully harnessed for in vitro differentiation, including liver, kidney and corneal tissue lineage cells. The incorporation of SS, in combination with biochemical and biophysical cues during MSC differentiation, may provide a promising avenue to improve the functionality of the differentiated cells by more closely mimicking the in vivo milieu.
Collapse
Affiliation(s)
- Seep Arora
- Department of Biomedical Engineering, National University of Singapore, 21 Lower Kent Ridge Rd, 117583, Singapore
| | - Akshaya Srinivasan
- Department of Biomedical Engineering, National University of Singapore, 21 Lower Kent Ridge Rd, 117583, Singapore
| | - Chak Ming Leung
- Department of Biomedical Engineering, National University of Singapore, 21 Lower Kent Ridge Rd, 117583, Singapore
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, 21 Lower Kent Ridge Rd, 117583, Singapore
| |
Collapse
|
28
|
Bartolotti I, Roseti L, Petretta M, Grigolo B, Desando G. A Roadmap of In Vitro Models in Osteoarthritis: A Focus on Their Biological Relevance in Regenerative Medicine. J Clin Med 2021; 10:1920. [PMID: 33925222 PMCID: PMC8124812 DOI: 10.3390/jcm10091920] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a multifaceted musculoskeletal disorder, with a high prevalence worldwide. Articular cartilage and synovial membrane are among the main biological targets in the OA microenvironment. Gaining more knowledge on the accuracy of preclinical in vitro OA models could open innovative avenues in regenerative medicine to bridge major gaps, especially in translation from animals to humans. Our methodological approach entailed searches on Scopus, the Web of Science Core Collection, and EMBASE databases to select the most relevant preclinical in vitro models for studying OA. Predicting the biological response of regenerative strategies requires developing relevant preclinical models able to mimic the OA milieu influencing tissue responses and organ complexity. In this light, standard 2D culture models lack critical properties beyond cell biology, while animal models suffer from several limitations due to species differences. In the literature, most of the in vitro models only recapitulate a tissue compartment, by providing fragmented results. Biotechnological advances may enable scientists to generate new in vitro models that combine easy manipulation and organ complexity. Here, we review the state-of-the-art of preclinical in vitro models in OA and outline how the different preclinical systems (inflammatory/biomechanical/microfluidic models) may be valid tools in regenerative medicine, describing their pros and cons. We then discuss the prospects of specific and combinatorial models to predict biological responses following regenerative approaches focusing on mesenchymal stromal cells (MSCs)-based therapies to reduce animal testing.
Collapse
Affiliation(s)
- Isabella Bartolotti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Livia Roseti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Mauro Petretta
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
- RegenHu Company, Z.I Du Vivier 22, 1690 Villaz-St-Pierre, Switzerland
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Giovanna Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| |
Collapse
|
29
|
Walker M, Luo J, Pringle EW, Cantini M. ChondroGELesis: Hydrogels to harness the chondrogenic potential of stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111822. [PMID: 33579465 DOI: 10.1016/j.msec.2020.111822] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 01/01/2023]
Abstract
The extracellular matrix is a highly complex microenvironment, whose various components converge to regulate cell fate. Hydrogels, as water-swollen polymer networks composed by synthetic or natural materials, are ideal candidates to create biologically active substrates that mimic these matrices and target cell behaviour for a desired tissue engineering application. Indeed, the ability to tune their mechanical, structural, and biochemical properties provides a framework to recapitulate native tissues. This review explores how hydrogels have been engineered to harness the chondrogenic response of stem cells for the repair of damaged cartilage tissue. The signalling processes involved in hydrogel-driven chondrogenesis are also discussed, identifying critical pathways that should be taken into account during hydrogel design.
Collapse
Affiliation(s)
- Matthew Walker
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK
| | - Jiajun Luo
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK
| | - Eonan William Pringle
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK
| | - Marco Cantini
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK.
| |
Collapse
|
30
|
Costa de Oliveira Souza CM, de Souza CF, Mogharbel BF, Irioda AC, Cavichiolo Franco CR, Sierakowski MR, Athayde Teixeira de Carvalho K. Nanostructured Cellulose-Gellan-Xyloglucan-Lysozyme Dressing Seeded with Mesenchymal Stem Cells for Deep Second-Degree Burn Treatment. Int J Nanomedicine 2021; 16:833-850. [PMID: 33584096 PMCID: PMC7875079 DOI: 10.2147/ijn.s289868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE In deep burns, wound contraction and hypertrophic scar formation can generate functional derangement and debilitation of the affected part. In order to improve the quality of healing in deep second-degree burns, we developed a new treatment in a preclinical model using nanostructured membranes seeded with mesenchymal stem cells (MSCs). METHODS Membranes were obtained by reconstitution of bacterial cellulose (reconstituted membrane [RM]) and produced by a dry-cast process, then RM was incorporated with 10% tamarind xyloglucan plus gellan gum 1:1 and 10% lysozyme (RMGT-LZ) and with 10% gellan gum and 10% lysozyme (RMG-LZ). Membrane hydrophobic/hydrophilic characteristics were investigated by static/dynamic contact-angle measurements. They were cultivated with MSCs, and cell adhesion, proliferation, and migration capacity was analyzed with MTT assays. Morphological and topographic characteristics were analyzed by scanning electron microscopy. MSC patterns in flow cytometry and differentiation into adipocytes and osteocytes were checked. In vivo assays used RMG-LZ and RMGT-LZ (with and without MSCs) in Rattus norvegicus rats submitted to burn protocol, and histological sections and collagen deposits were analyzed and immunocytochemistry assay performed. RESULTS In vitro results demonstrated carboxyl and amine groups made the membranes moderately hydrophobic and xyloglucan inclusion decreased wettability, favoring MSC adhesion, proliferation, and differentiation. In vivo, we obtained 40% and 60% reduction in acute/chronic inflammatory infiltrates, 96% decrease in injury area, increased vascular proliferation and collagen deposition, and complete epithelialization after 30 days. MSCs were detected in burned tissue, confirming they had homed and proliferated in vivo. CONCLUSION Nanostructured cellulose-gellan-xyloglucan-lysozyme dressings, especially when seeded with MSCs, improved deep second-degree burn regeneration.
Collapse
Affiliation(s)
- Carolina Maria Costa de Oliveira Souza
- Stem Cell Research Laboratory, Cell Therapy and Biotechnology in Regenerative Medicine Department, Pequeno Príncipe Faculties and the Pelé Pequeno Príncipe Research Institute, Curitiba, Paraná, Brazil
| | - Clayton Fernandes de Souza
- Chemistry Undergraduate Program, School of Education and Humanities, Pontifícia Universidade Católica Do Paraná, Curitiba, Paraná, Brazil
- BioPol, Chemistry Department, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Bassam Felipe Mogharbel
- Stem Cell Research Laboratory, Cell Therapy and Biotechnology in Regenerative Medicine Department, Pequeno Príncipe Faculties and the Pelé Pequeno Príncipe Research Institute, Curitiba, Paraná, Brazil
| | - Ana Carolina Irioda
- Stem Cell Research Laboratory, Cell Therapy and Biotechnology in Regenerative Medicine Department, Pequeno Príncipe Faculties and the Pelé Pequeno Príncipe Research Institute, Curitiba, Paraná, Brazil
| | | | | | - Katherine Athayde Teixeira de Carvalho
- Stem Cell Research Laboratory, Cell Therapy and Biotechnology in Regenerative Medicine Department, Pequeno Príncipe Faculties and the Pelé Pequeno Príncipe Research Institute, Curitiba, Paraná, Brazil
| |
Collapse
|
31
|
Bicer M, Cottrell GS, Widera D. Impact of 3D cell culture on bone regeneration potential of mesenchymal stromal cells. Stem Cell Res Ther 2021; 12:31. [PMID: 33413646 PMCID: PMC7791873 DOI: 10.1186/s13287-020-02094-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022] Open
Abstract
As populations age across the world, osteoporosis and osteoporosis-related fractures are becoming the most prevalent degenerative bone diseases. More than 75 million patients suffer from osteoporosis in the USA, the EU and Japan. Furthermore, it is anticipated that the number of patients affected by osteoporosis will increase by a third by 2050. Although conventional therapies including bisphosphonates, calcitonin and oestrogen-like drugs can be used to treat degenerative diseases of the bone, they are often associated with serious side effects including the development of oesophageal cancer, ocular inflammation, severe musculoskeletal pain and osteonecrosis of the jaw.The use of autologous mesenchymal stromal cells/mesenchymal stem cells (MSCs) is a possible alternative therapeutic approach to tackle osteoporosis while overcoming the limitations of traditional treatment options. However, osteoporosis can cause a decrease in the numbers of MSCs, induce their senescence and lower their osteogenic differentiation potential.Three-dimensional (3D) cell culture is an emerging technology that allows a more physiological expansion and differentiation of stem cells compared to cultivation on conventional flat systems.This review will discuss current understanding of the effects of different 3D cell culture systems on proliferation, viability and osteogenic differentiation, as well as on the immunomodulatory and anti-inflammatory potential of MSCs.
Collapse
Affiliation(s)
- Mesude Bicer
- Stem Cell Biology and Regenerative Medicine Group, Reading School of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, RG6 6AP, UK
| | - Graeme S Cottrell
- Cellular and Molecular Neuroscience, School of Pharmacy, University of Reading, Reading, UK
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, Reading School of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, RG6 6AP, UK.
| |
Collapse
|
32
|
Advanced 3D Cell Culture Techniques in Micro-Bioreactors, Part II: Systems and Applications. Processes (Basel) 2020. [DOI: 10.3390/pr9010021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this second part of our systematic review on the research area of 3D cell culture in micro-bioreactors we give a detailed description of the published work with regard to the existing micro-bioreactor types and their applications, and highlight important results gathered with the respective systems. As an interesting detail, we found that micro-bioreactors have already been used in SARS-CoV research prior to the SARS-CoV2 pandemic. As our literature research revealed a variety of 3D cell culture configurations in the examined bioreactor systems, we defined in review part one “complexity levels” by means of the corresponding 3D cell culture techniques applied in the systems. The definition of the complexity is thereby based on the knowledge that the spatial distribution of cell-extracellular matrix interactions and the spatial distribution of homologous and heterologous cell–cell contacts play an important role in modulating cell functions. Because at least one of these parameters can be assigned to the 3D cell culture techniques discussed in the present review, we structured the studies according to the complexity levels applied in the MBR systems.
Collapse
|
33
|
Electrical Stimulation of Adipose-Derived Stem Cells in 3D Nanofibrillar Cellulose Increases Their Osteogenic Potential. Biomolecules 2020; 10:biom10121696. [PMID: 33353222 PMCID: PMC7766661 DOI: 10.3390/biom10121696] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Due to the ageing population, there is a steadily increasing incidence of osteoporosis and osteoporotic fractures. As conventional pharmacological therapy options for osteoporosis are often associated with severe side effects, bone grafts are still considered the clinical gold standard. However, the availability of viable, autologous bone grafts is limited making alternative cell-based strategies a promising therapeutic alternative. Adipose-derived stem cells (ASCs) are a readily available population of mesenchymal stem/stromal cells (MSCs) that can be isolated within minimally invasive surgery. This ease of availability and their ability to undergo osteogenic differentiation makes ASCs promising candidates for cell-based therapies for bone fractures. Recent studies have suggested that both exposure to electrical fields and cultivation in 3D can positively affect osteogenic potential of MSCs. To elucidate the osteoinductive potential of a combination of these biophysical cues on ASCs, cells were embedded within anionic nanofibrillar cellulose (aNFC) hydrogels and exposed to electrical stimulation (ES) for up to 21 days. ES was applied to ASCs in 2D and 3D at a voltage of 0.1 V/cm with a duration of 0.04 ms, and a frequency of 10 Hz for 30 min per day. Exposure of ASCs to ES in 3D resulted in high alkaline phosphatase (ALP) activity and in an increased mineralisation evidenced by Alizarin Red S staining. Moreover, ES in 3D aNFC led to an increased expression of the osteogenic markers osteopontin and osteocalcin and a rearrangement and alignment of the actin cytoskeleton. Taken together, our data suggest that a combination of ES with 3D cell culture can increase the osteogenic potential of ASCs. Thus, exposure of ASCs to these biophysical cues might improve the clinical outcomes of regenerative therapies in treatment of osteoporotic fractures.
Collapse
|
34
|
Advanced 3D Cell Culture Techniques in Micro-Bioreactors, Part I: A Systematic Analysis of the Literature Published between 2000 and 2020. Processes (Basel) 2020. [DOI: 10.3390/pr8121656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bioreactors have proven useful for a vast amount of applications. Besides classical large-scale bioreactors and fermenters for prokaryotic and eukaryotic organisms, micro-bioreactors, as specialized bioreactor systems, have become an invaluable tool for mammalian 3D cell cultures. In this systematic review we analyze the literature in the field of eukaryotic 3D cell culture in micro-bioreactors within the last 20 years. For this, we define complexity levels with regard to the cellular 3D microenvironment concerning cell–matrix-contact, cell–cell-contact and the number of different cell types present at the same time. Moreover, we examine the data with regard to the micro-bioreactor design including mode of cell stimulation/nutrient supply and materials used for the micro-bioreactors, the corresponding 3D cell culture techniques and the related cellular microenvironment, the cell types and in vitro models used. As a data source we used the National Library of Medicine and analyzed the studies published from 2000 to 2020.
Collapse
|
35
|
Bonetti L, De Nardo L, Farè S. Thermo-Responsive Methylcellulose Hydrogels: From Design to Applications as Smart Biomaterials. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:486-513. [DOI: 10.1089/ten.teb.2020.0202] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Lorenzo Bonetti
- Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Milan, Italy
| | - Luigi De Nardo
- Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Milan, Italy
- INSTM, National Interuniversity Consortium of Materials Science and Technology, Florence, Italy
| | - Silvia Farè
- Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Milan, Italy
- INSTM, National Interuniversity Consortium of Materials Science and Technology, Florence, Italy
| |
Collapse
|
36
|
Hogan KJ, Mikos AG. Biodegradable thermoresponsive polymers: Applications in drug delivery and tissue engineering. POLYMER 2020. [DOI: 10.1016/j.polymer.2020.123063] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
37
|
Mokhtarinia K, Masaeli E. Transiently thermally responsive surfaces: Concepts for cell sheet engineering. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.110076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
38
|
Bonifacio MA, Cochis A, Cometa S, Gentile P, Scalzone A, Scalia AC, Rimondini L, De Giglio E. From the sea to the bee: Gellan gum-honey-diatom composite to deliver resveratrol for cartilage regeneration under oxidative stress conditions. Carbohydr Polym 2020; 245:116410. [DOI: 10.1016/j.carbpol.2020.116410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/03/2020] [Accepted: 05/03/2020] [Indexed: 01/22/2023]
|
39
|
Frazier T, Alarcon A, Wu X, Mohiuddin OA, Motherwell JM, Carlsson AH, Christy RJ, Edwards JV, Mackin RT, Prevost N, Gloster E, Zhang Q, Wang G, Hayes DJ, Gimble JM. Clinical Translational Potential in Skin Wound Regeneration for Adipose-Derived, Blood-Derived, and Cellulose Materials: Cells, Exosomes, and Hydrogels. Biomolecules 2020; 10:E1373. [PMID: 32992554 PMCID: PMC7650547 DOI: 10.3390/biom10101373] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
Acute and chronic skin wounds due to burns, pressure injuries, and trauma represent a substantial challenge to healthcare delivery with particular impacts on geriatric, paraplegic, and quadriplegic demographics worldwide. Nevertheless, the current standard of care relies extensively on preventive measures to mitigate pressure injury, surgical debridement, skin flap procedures, and negative pressure wound vacuum measures. This article highlights the potential of adipose-, blood-, and cellulose-derived products (cells, decellularized matrices and scaffolds, and exosome and secretome factors) as a means to address this unmet medical need. The current status of this research area is evaluated and discussed in the context of promising avenues for future discovery.
Collapse
Affiliation(s)
- Trivia Frazier
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
| | - Andrea Alarcon
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
| | - Xiying Wu
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
- LaCell LLC, New Orleans, LA 70148, USA
| | - Omair A. Mohiuddin
- Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Science, University of Karachi, Karachi 75270, Pakistan;
| | | | - Anders H. Carlsson
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX 78234, USA; (A.H.C.); (R.J.C.)
| | - Robert J. Christy
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX 78234, USA; (A.H.C.); (R.J.C.)
| | - Judson V. Edwards
- Southern Regional Research Center-USDA-ARS, New Orleans, LA 70124, USA; (J.V.E.); (R.T.M.); (N.P.)
| | - Robert T. Mackin
- Southern Regional Research Center-USDA-ARS, New Orleans, LA 70124, USA; (J.V.E.); (R.T.M.); (N.P.)
| | - Nicolette Prevost
- Southern Regional Research Center-USDA-ARS, New Orleans, LA 70124, USA; (J.V.E.); (R.T.M.); (N.P.)
| | - Elena Gloster
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA; (E.G.); (Q.Z.); (G.W.)
| | - Qiang Zhang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA; (E.G.); (Q.Z.); (G.W.)
| | - Guangdi Wang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA; (E.G.); (Q.Z.); (G.W.)
| | - Daniel J. Hayes
- Department of Biomedical Engineering, State College, Pennsylvania State University, Centre County, PA 16802, USA;
| | - Jeffrey M. Gimble
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
- LaCell LLC, New Orleans, LA 70148, USA
| |
Collapse
|
40
|
Cartilage repair using stem cells & biomaterials: advancement from bench to bedside. Mol Biol Rep 2020; 47:8007-8021. [PMID: 32888123 DOI: 10.1007/s11033-020-05748-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 08/28/2020] [Indexed: 10/23/2022]
Abstract
Osteoarthritis (OA) involves gradual destruction of articular cartilagemanifested by pain, stiffness of joints, and impaired movement especially in knees and hips. Non-vascularity of this tissue hinders its self-regenerative capacity and thus, the application of reparative or restorative modalities becomes imperative in OA treatment. In recent years, stem cell-based therapies have been explored as potential modalities for addressing OA complications. While mesenchymal stem cells (MSCs) hold immense promise, the recapitulation of native articular cartilage usingMSCs remains elusive. In this review, we have highlighted the chondrogenic potential of MSCs, factors guiding in vitro chondrogenic differentiation, biomaterials available for cartilage repair, their current market status, and the outcomes of major clinical trials. Our search on ClinicalTrials.gov using terms "stem cell" and "osteoarthritis" yielded 83 results. An analysis of the 29 trials that have been completed revealed differences in source of MSCs (bone marrow, adipose tissue, umbilical cord etc.), cell type (autologous or allogenic), and dose administered. Moreover, only 02 out of 29 studies have reported the use of matrix for cartilage repair. From future perspective, aconsensus on choice of cells, differentiation inducers, biomaterials, and clinical settings might pave a way for concocting robust strategies to improve the clinical applicability of biomimetic neocartilage constructs.
Collapse
|
41
|
Sory DR, Amin HD, Chapman DJ, Proud WG, Rankin SM. Replicating landmine blast loading in cellular in vitro models. Phys Biol 2020; 17:056001. [PMID: 32141440 DOI: 10.1088/1478-3975/ab7d1c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Trauma arising from landmines and improvised explosive devices promotes heterotopic ossification, the formation of extra-skeletal bone in non-osseous tissue. To date, experimental platforms that can replicate the loading parameter space relevant to improvised explosive device and landmine blast wave exposure have not been available to study the effects of such non-physiological mechanical loading on cells. Here, we present the design and calibration of three distinct in vitro experimental loading platforms that allow us to replicate the spectrum of loading conditions recorded in near-field blast wave exposure. We subjected cells in suspension or in a three-dimensional hydrogel to strain rates up to 6000 s-1 and pressure levels up to 45 MPa. Our results highlight that cellular activation is regulated in a non-linear fashion-not by a single mechanical parameter, it is the combined action of the applied mechanical pressure, rate of loading and loading impulse, along with the extracellular environment used to convey the pressure waves. Finally, our research indicates that PO MSCs are finely tuned to respond to mechanical stimuli that fall within defined ranges of loading.
Collapse
Affiliation(s)
- David R Sory
- Institute of Shock Physics, Department of Physics, Imperial College London, London SW7 2AZ, United Kingdom. National Heart & Lung Institute, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom. The Royal British Legion-Centre for Blast Injury Studies, Imperial College London, London SW7 2AZ, United Kingdom
| | | | | | | | | |
Collapse
|
42
|
Najmi Z, Kumar A, Scalia AC, Cochis A, Obradovic B, Grassi FA, Leigheb M, Lamghari M, Loinaz I, Gracia R, Rimondini L. Evaluation of Nisin and LL-37 Antimicrobial Peptides as Tool to Preserve Articular Cartilage Healing in a Septic Environment. Front Bioeng Biotechnol 2020; 8:561. [PMID: 32596225 PMCID: PMC7304409 DOI: 10.3389/fbioe.2020.00561] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022] Open
Abstract
Cartilage repair still represents a challenge for clinicians and only few effective therapies are nowadays available. In fact, surgery is limited by the tissue poor self-healing capacity while the autologous transplantation is often forsaken due to the poor in vitro expansion capacity of chondrocytes. Biomaterials science offers a unique alternative based on the replacement of the injured tissue with an artificial tissue-mimicking scaffold. However, the implantation surgical practices and the scaffold itself can be a source of bacterial infection that currently represents the first reason of implants failure due to the increasing antibiotics resistance of pathogens. So, alternative antibacterial tools to prevent infections and consequent device removal are urgently required. In this work, the role of Nisin and LL-37 peptides has been investigated as alternative to antibiotics to their antimicrobial performances for direct application at the surgical site or as doping chemicals for devices aimed at articular cartilage repair. First, peptides cytocompatibility was investigated toward human mesenchymal stem cells to determine safe concentrations; then, the broad-range antibacterial activity was verified toward the Gram-positive Staphylococcus aureus and Staphylococcus epidermidis as well as the Gram-negative Escherichia coli and Aggregatibacter actinomycetemcomitans pathogens. The peptides selective antibacterial activity was verified by a cells-bacteria co-culture assay, while chondrogenesis was assayed to exclude any interference within the differentiation route to simulate the tissue repair. In the next phase, the experiments were repeated by moving from the cell monolayer model to 3D cartilage-like spheroids to revisit the peptides activity in a more physiologically relevant environment model. Finally, the spheroid model was applied in a perfusion bioreactor to simulate an infection in the presence of circulating peptides within a physiological environment. Results suggested that 75 μg/ml Nisin can be considered as a very promising candidate since it was shown to be more cytocompatible and potent against the investigated bacteria than LL-37 in all the tested models.
Collapse
Affiliation(s)
- Ziba Najmi
- Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Novara, Italy
| | - Ajay Kumar
- Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Novara, Italy
| | - Alessandro C Scalia
- Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Novara, Italy
| | - Andrea Cochis
- Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Novara, Italy
| | - Bojana Obradovic
- Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Federico A Grassi
- Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy
| | - Massimiliano Leigheb
- Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy
| | - Meriem Lamghari
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Iraida Loinaz
- CIDETEC Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Raquel Gracia
- CIDETEC Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Lia Rimondini
- Department of Health Sciences, University of Piemonte Orientale UPO, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Novara, Italy
| |
Collapse
|
43
|
Kim H, Yi JY, Kim BG, Song JE, Jeong HJ, Kim HR. Development of cellulose-based conductive fabrics with electrical conductivity and flexibility. PLoS One 2020; 15:e0233952. [PMID: 32498075 PMCID: PMC7272206 DOI: 10.1371/journal.pone.0233952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/11/2020] [Indexed: 11/18/2022] Open
Abstract
This study aimed to produce cellulose-based conductive fabrics with electrical conductivity and flexibility. Bacterial cellulose (BC) and three chemical cellulose (CC), namely methyl cellulose (MC), hydroxypropyl cellulose (HPMC) and carboxymethyl cellulose (CMC) were in situ polymerized with aniline and the four conductive cellulose fabrics were compared and evaluated. Matrix-assisted laser desorption/ionization time-of-flight mass spectroscopy analysis confirmed that three CC-PANI composites displayed longer and more stable polymerization pattern than BC-PANI because of the different polymerization method: bulk polymerization for BC-PANI and emulsion polymerization for CC-PANI, respectively. The electrical conductivity of BC-PANI and CC-PANI were ranging from 0.962 × 10-2 S/cm to 2.840 × 10-2 S/cm. MC-PANI showed the highest electrical conductivity among the four conductive cellulose fabrics. The flexibility and crease recovery results showed that MC-PANI had the highest flexibility compared to BC-PANI, HPMC-PANI, and CMC-PANI. These results have confirmed that the electrical conductivity and flexibility were influenced by the type of cellulose, and MC-PANI was found to have the best performance in the electrical conductivity and flexibility.
Collapse
Affiliation(s)
- Hyunjin Kim
- Department of Clothing and Textiles, Sookmyung Women's University, Seoul, South Korea
| | - Joon-Yeop Yi
- Interdisciplinary Program of Bioengineering, Seoul National University, Seoul, South Korea
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
| | - Byung-Gee Kim
- Interdisciplinary Program of Bioengineering, Seoul National University, Seoul, South Korea
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul, South Korea
| | - Ji Eun Song
- Human Convergence Technology Group, Korea Institute of Industrial Technology, Ansan, South Korea
| | - Hee-Jin Jeong
- Department of Biological and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Hye Rim Kim
- Department of Clothing and Textiles, Sookmyung Women's University, Seoul, South Korea
| |
Collapse
|
44
|
Xu Y, Xu Y, Bi B, Hou M, Yao L, Du Q, He A, Liu Y, Miao C, Liang X, Jiang X, Zhou G, Cao Y. A moldable thermosensitive hydroxypropyl chitin hydrogel for 3D cartilage regeneration in vitro and in vivo. Acta Biomater 2020; 108:87-96. [PMID: 32268237 DOI: 10.1016/j.actbio.2020.03.039] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 03/18/2020] [Accepted: 03/27/2020] [Indexed: 12/21/2022]
Abstract
Because of poor self-repair capacity, the repair of cartilage defect is always a great challenge in clinical treatment. In vitro cartilage regeneration provides a potential strategy for functional reconstruction of cartilage defect. Hydrogel has been known as an ideal cartilage regeneration scaffold. However, to date, in vitro cartilage regeneration based on hydrogel has not achieved satisfactory results. The current study explored the feasibility of in vitro 3D cartilage regeneration based on a moldable thermosensitive hydroxypropyl chitin (HPCH) hydrogel and its in vivo fate. The thermosensitive HPCH hydrogel was prepared and characterized. Goat auricular chondrocytes were encapsulated into the HPCH hydrogel to form a chondrocyte-hydrogel construct. The constructs were injected subcutaneously into nude mice or molded into different shapes for in vitro chondrogenic culture followed by in vivo implantation. The results demonstrated that the HPCH hydrogel possessed satisfactory gelation properties (gelation time < 18 s at 37 °C), biocompatibility (cell amount almost doubled within one week), and the ability to be applied as an injectable hydrogel for cartilage regeneration. All the constructs of in vitro culture basically maintained their original shapes (in vitro to initial: 110.8%) and displayed typical cartilaginous features with abundant lacunae and cartilage specific matrix deposition. These in vitro samples became more mature with prolonged in vivo implantation and largely maintained the original shape (in vivo to in vitro: 103.5%). These results suggested that the moldable thermosensitive HPCH hydrogel can serve as a promising scaffold for cartilage regeneration with defined shapes in vitro and in vivo. STATEMENT OF SIGNIFICANCE: Because of avascular and non-nervous characteristic of cartilage, in vitro regeneration plays an important role in reconstructing cartilage function. Hydrogel has been known as an ideal cartilage regeneration scaffold. However, to date, in vitro cartilage regeneration based on hydrogel has not achieved satisfactory results. The current study demonstrated that the chondrocyte-hydrogel construct generated by high density of chondrocytes encapsulated into a thermosensitive HPCH hydrogel could successfully regenerate in vitro typical cartilage-like tissue with defined shapes and further mature to form homogeneous cartilage with their original shapes after in vivo implantation. The current study indicated that the moldable thermosensitive HPCH hydrogel could serve as a promising scaffold for in vitro and in vivo cartilage regeneration with different shapes.
Collapse
|
45
|
New Insights on Mechanical Stimulation of Mesenchymal Stem Cells for Cartilage Regeneration. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10082927] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Successful tissue regeneration therapies require further understanding of the environment in which the cells are destined to be set. The aim is to structure approaches that aspire to a holistic view of biological systems and to scientific reliability. Mesenchymal stem cells represent a valuable resource for cartilage tissue engineering, due to their chondrogenic differentiation capacity. Promoting chondrogenesis, not only by growth factors but also by exogenous enhancers such as biomechanics, represents a technical enhancement. Tribological evaluation of the articular joint has demonstrated how mechanical stimuli play a pivotal role in cartilage repair and participate in the homeostasis of this tissue. Loading stresses, physiologically experienced by chondrocytes, can upregulate the production of proteins like glycosaminoglycan or collagen, fundamental for articular wellness, as well as promote and preserve cell viability. Therefore, there is a rising interest in the development of bioreactor devices that impose compression, shear stress, and hydrostatic pressure on stem cells. This strategy aims to mimic chondrogenesis and overcome complications like hypertrophic phenotyping and inappropriate mechanical features. This review will analyze the dynamics inside the joint, the natural stimuli experienced by the chondrocytes, and how the biomechanical stimuli can be applied to a stem cell culture in order to induce chondrogenesis.
Collapse
|
46
|
Advances in cartilage repair: The influence of inorganic clays to improve mechanical and healing properties of antibacterial Gellan gum-Manuka honey hydrogels. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 108:110444. [DOI: 10.1016/j.msec.2019.110444] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/31/2019] [Accepted: 11/15/2019] [Indexed: 12/15/2022]
|
47
|
Spicer CD. Hydrogel scaffolds for tissue engineering: the importance of polymer choice. Polym Chem 2020; 11:184-219. [DOI: 10.1039/c9py01021a] [Citation(s) in RCA: 275] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
We explore the design and synthesis of hydrogel scaffolds for tissue engineering from the perspective of the underlying polymer chemistry. The key polymers, properties and architectures used, and their effect on tissue growth are discussed.
Collapse
|
48
|
Gholamali I. Stimuli-Responsive Polysaccharide Hydrogels for Biomedical Applications: a Review. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00134-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
49
|
Del Bakhshayesh AR, Asadi N, Alihemmati A, Tayefi Nasrabadi H, Montaseri A, Davaran S, Saghati S, Akbarzadeh A, Abedelahi A. An overview of advanced biocompatible and biomimetic materials for creation of replacement structures in the musculoskeletal systems: focusing on cartilage tissue engineering. J Biol Eng 2019; 13:85. [PMID: 31754372 PMCID: PMC6854707 DOI: 10.1186/s13036-019-0209-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/23/2019] [Indexed: 01/06/2023] Open
Abstract
Tissue engineering, as an interdisciplinary approach, is seeking to create tissues with optimal performance for clinical applications. Various factors, including cells, biomaterials, cell or tissue culture conditions and signaling molecules such as growth factors, play a vital role in the engineering of tissues. In vivo microenvironment of cells imposes complex and specific stimuli on the cells, and has a direct effect on cellular behavior, including proliferation, differentiation and extracellular matrix (ECM) assembly. Therefore, to create appropriate tissues, the conditions of the natural environment around the cells should be well imitated. Therefore, researchers are trying to develop biomimetic scaffolds that can produce appropriate cellular responses. To achieve this, we need to know enough about biomimetic materials. Scaffolds made of biomaterials in musculoskeletal tissue engineering should also be multifunctional in order to be able to function better in mechanical properties, cell signaling and cell adhesion. Multiple combinations of different biomaterials are used to improve above-mentioned properties of various biomaterials and to better imitate the natural features of musculoskeletal tissue in the culture medium. These improvements ultimately lead to the creation of replacement structures in the musculoskeletal system, which are closer to natural tissues in terms of appearance and function. The present review article is focused on biocompatible and biomimetic materials, which are used in musculoskeletal tissue engineering, in particular, cartilage tissue engineering.
Collapse
Affiliation(s)
- Azizeh Rahmani Del Bakhshayesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahideh Asadi
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Alihemmati
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Tayefi Nasrabadi
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azadeh Montaseri
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Saghati
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Akbarzadeh
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
50
|
Optically Transparent Anionic Nanofibrillar Cellulose Is Cytocompatible with Human Adipose Tissue-Derived Stem Cells and Allows Simple Imaging in 3D. Stem Cells Int 2019; 2019:3106929. [PMID: 31687032 PMCID: PMC6800951 DOI: 10.1155/2019/3106929] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
The anti-inflammatory and immunomodulatory properties of human mesenchymal stromal cells (MSCs) are a focus within regenerative medicine. However, 2D cultivation of MSCs for extended periods results in abnormal cell polarity, chromosomal changes, reduction in viability, and altered differentiation potential. As an alternative, various 3D hydrogels have been developed which mimic the endogenous niche of MSCs. Nevertheless, imaging cells embedded within 3D hydrogels often suffers from low signal-to-noise ratios which can be at least partly attributed to the high light absorbance and light scattering of the hydrogels in the visible light spectrum. In this study, human adipose tissue-derived MSCs (ADSCs) are cultivated within an anionic nanofibrillar cellulose (aNFC) hydrogel. It is demonstrated that aNFC forms nanofibres arranged as a porous network with low light absorbance in the visible spectrum. Moreover, it is shown that aNFC is cytocompatible, allowing for MSC proliferation, maintaining cell viability and multilineage differentiation potential. Finally, aNFC is compatible with scanning electron microscopy (SEM) and light microscopy including the application of conventional dyes, fluorescent probes, indirect immunocytochemistry, and calcium imaging. Overall, the results indicate that aNFC represents a promising 3D material for the expansion of MSCs whilst allowing detailed examination of cell morphology and cellular behaviour.
Collapse
|