1
|
Han R, Luo L, Wei C, Qiao Y, Xie J, Pan X, Xing J. Stiffness-tunable biomaterials provide a good extracellular matrix environment for axon growth and regeneration. Neural Regen Res 2025; 20:1364-1376. [PMID: 39075897 PMCID: PMC11624885 DOI: 10.4103/nrr.nrr-d-23-01874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/31/2024] [Accepted: 03/16/2024] [Indexed: 07/31/2024] Open
Abstract
Neuronal growth, extension, branching, and formation of neural networks are markedly influenced by the extracellular matrix-a complex network composed of proteins and carbohydrates secreted by cells. In addition to providing physical support for cells, the extracellular matrix also conveys critical mechanical stiffness cues. During the development of the nervous system, extracellular matrix stiffness plays a central role in guiding neuronal growth, particularly in the context of axonal extension, which is crucial for the formation of neural networks. In neural tissue engineering, manipulation of biomaterial stiffness is a promising strategy to provide a permissive environment for the repair and regeneration of injured nervous tissue. Recent research has fine-tuned synthetic biomaterials to fabricate scaffolds that closely replicate the stiffness profiles observed in the nervous system. In this review, we highlight the molecular mechanisms by which extracellular matrix stiffness regulates axonal growth and regeneration. We highlight the progress made in the development of stiffness-tunable biomaterials to emulate in vivo extracellular matrix environments, with an emphasis on their application in neural repair and regeneration, along with a discussion of the current limitations and future prospects. The exploration and optimization of the stiffness-tunable biomaterials has the potential to markedly advance the development of neural tissue engineering.
Collapse
Affiliation(s)
- Ronglin Han
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Lanxin Luo
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Caiyan Wei
- Department of Medicinal Chemistry, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yaru Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jiming Xie
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xianchao Pan
- Department of Medicinal Chemistry, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Juan Xing
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
2
|
Torresan V, Dedroog LM, Deschaume O, Koos E, Lettinga MP, Gandin A, Pelosin M, Zanconato F, Brusatin G, Bartic C. Nanocellulose-collagen composites as advanced biomaterials for 3D in-vitro neuronal model systems. Carbohydr Polym 2025; 348:122901. [PMID: 39567136 DOI: 10.1016/j.carbpol.2024.122901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/14/2024] [Accepted: 10/20/2024] [Indexed: 11/22/2024]
Abstract
Studying brain diseases and developing therapies requires versatile in vitro systems for long-term neuronal cultures. SH-SY5Y neuroblastoma cells are ideal for modeling neurodegenerative diseases. Although SH-SY5Y cells are commonly used in 2D cultures, 3D systems offer more physiologically relevant models. Studies have shown 3D culturing up to 7 days, but a simple, reproducible, and tunable system has yet to be identified. Cellulose holds potential to fulfill these needs. Cellulose and its derivatives are sustainable, cytocompatible, and ideal for synthesizing biocompatible hydrogels. Its abundance and ease of chemical modification make it a highly attractive biomaterial. This study explored nanocellulose-based hydrogels for promoting neuronal growth and morphogenesis. To enhance cell adhesion, a small amount of collagen was added to the hydrogel, and the resulting cell morphologies were analyzed and compared with those cultured in collagen and Matrigel. By chemically oxidizing cellulose and adjusting the blend, we developed composites that maintained neuronal viability for over 14 days in 3D cultures. Our findings show that nanocellulose-collagen composites offer superior cytocompatibility, promoting neuronal viability and neurite outgrowth more effectively than Matrigel and collagen. These tunable biomaterials support long-term 3D neuronal cultures, making them valuable for creating standardized models for disease research and drug development.
Collapse
Affiliation(s)
- Veronica Torresan
- Department of Industrial Engineering, University of Padova and INSTM, via Marzolo 9, 35131 Padova, Italy
| | - Lens Martijn Dedroog
- Soft Matter Physics and Biophysics Unit, Department of Physics and Astronomy, KU Leuven, 3001 Leuven, Belgium
| | - Olivier Deschaume
- Soft Matter Physics and Biophysics Unit, Department of Physics and Astronomy, KU Leuven, 3001 Leuven, Belgium
| | - Erin Koos
- Soft Matter, Rheology and Technology, Department of Chemical Engineering, KU Leuven, 3001 Leuven, Belgium
| | - Minne Paul Lettinga
- Soft Matter Physics and Biophysics Unit, Department of Physics and Astronomy, KU Leuven, 3001 Leuven, Belgium; Biological Information Processing IBI-4, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Alessandro Gandin
- Department of Industrial Engineering, University of Padova and INSTM, via Marzolo 9, 35131 Padova, Italy
| | - Margherita Pelosin
- Department of Industrial Engineering, University of Padova and INSTM, via Marzolo 9, 35131 Padova, Italy
| | - Francesca Zanconato
- Department of Molecular Medicine, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Giovanna Brusatin
- Department of Industrial Engineering, University of Padova and INSTM, via Marzolo 9, 35131 Padova, Italy.
| | - Carmen Bartic
- Soft Matter Physics and Biophysics Unit, Department of Physics and Astronomy, KU Leuven, 3001 Leuven, Belgium.
| |
Collapse
|
3
|
Li J, Hietel B, Brunk MGK, Reimers A, Willems C, Groth T, Cynis H, Adelung R, Schütt F, Sacher WD, Poon JKS. 3D-printed microstructured alginate scaffolds for neural tissue engineering. Trends Biotechnol 2024:S0167-7799(24)00304-4. [PMID: 39658448 DOI: 10.1016/j.tibtech.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/18/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024]
Abstract
Alginate (Alg) is a versatile biopolymer for scaffold engineering and a bioink component widely used for direct cell printing. However, due to a lack of intrinsic cell-binding sites, Alg must be functionalized for cellular adhesion when used as a scaffold. Moreover, direct cell-laden ink 3D printing requires tedious disinfection procedures and cell viability is compromised by shear stress. Here, we demonstrate proof-of-concept, bioactive additive-free, microstructured Alg (M-Alg) scaffolds for neuron culture. The M-Alg scaffold was formed by introducing tetrapod-shaped ZnO (t-ZnO) microparticles into the ink as structural templates for interconnected channels and textured surfaces in the 3D-printed Alg scaffold, which were subsequently removed. Neurons exhibited significantly improved adhesion and growth on these M-Alg scaffolds compared with pristine Alg (P-Alg) scaffolds, with extensive neurite outgrowth and spontaneous neural activity, indicating the maturation of neuronal networks. These transparent, porous, additive-free Alg-based scaffolds with neuron affinity are promising for neuroregenerative and organoid-related research.
Collapse
Affiliation(s)
- Jianfeng Li
- Max Planck Institute of Microstructure Physics, Weinberg 2, Halle, 06120, Germany; Max Planck-University of Toronto Centre for Neural Science and Technology, Toronto, Canada.
| | - Benjamin Hietel
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Weinbergweg 22, 06120, Halle, Germany
| | - Michael G K Brunk
- Max Planck Institute of Microstructure Physics, Weinberg 2, Halle, 06120, Germany; Max Planck-University of Toronto Centre for Neural Science and Technology, Toronto, Canada
| | - Armin Reimers
- Functional Nanomaterials, Department of Materials Science, Kiel University, Kaiserstraße 2, 24143 Kiel, Germany
| | - Christian Willems
- Department of Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120, Halle, Germany
| | - Thomas Groth
- Department of Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120, Halle, Germany
| | - Holger Cynis
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Weinbergweg 22, 06120, Halle, Germany; Junior Research Group, Immunomodulation in Pathophysiological Processes, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120, Halle, Germany
| | - Rainer Adelung
- Functional Nanomaterials, Department of Materials Science, Kiel University, Kaiserstraße 2, 24143 Kiel, Germany
| | - Fabian Schütt
- Functional Nanomaterials, Department of Materials Science, Kiel University, Kaiserstraße 2, 24143 Kiel, Germany
| | - Wesley D Sacher
- Max Planck Institute of Microstructure Physics, Weinberg 2, Halle, 06120, Germany; Max Planck-University of Toronto Centre for Neural Science and Technology, Toronto, Canada
| | - Joyce K S Poon
- Max Planck Institute of Microstructure Physics, Weinberg 2, Halle, 06120, Germany; Max Planck-University of Toronto Centre for Neural Science and Technology, Toronto, Canada; Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Road, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Gould ML, Downes NJ, Woolley AG, Hussaini HM, Ratnayake JT, Ali MA, Friedlander LT, Cooper PR. Harnessing the Regenerative Potential of Purified Bovine Dental Pulp and Dentin Extracellular Matrices in a Chitosan/Alginate Hydrogel. Macromol Biosci 2024; 24:e2400254. [PMID: 38938070 DOI: 10.1002/mabi.202400254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/24/2024] [Indexed: 06/29/2024]
Abstract
When a tooth is diseased or damaged through caries, bioactive molecules are liberated from the pulp and dentin as part of the natural response to injury and these are key molecules for stimulating stem cell responses for tissue repair. Incorporation of these extracellular-matrix (ECM)-derived molecules into a hydrogel model can mimic in vivo conditions to enable dentin-pulp complex regeneration. Here, a chitosan/alginate (C/A) hydrogel is developed to sequester bovine ECM extracts. Human dental pulp cells (hDPCs) are cultured with these constructs and proliferation and cytotoxicity assays confirm that these C/A hydrogels are bioactive. Sequential z-axis fluorescent imaging visualizes hDPCs protruding into the hydrogel as it degraded. Alizarin red S staining shows that hDPCs cultured with the hydrogels display increased calcium-ion deposition, with dentin ECM stimulating the highest levels. Alkaline phosphatase activity is increased, as is expression of transforming growth factor-beta as demonstrated using immunocytochemistry. Directional analysis following phase contrast kinetic image capture demonstrates that both dentin and pulp ECM molecules act as chemoattractants for hDPCs. Data from this study demonstrate that purified ECM from dental pulp and dentin when delivered in a C/A hydrogel stimulates dental tissue repair processes in vitro.
Collapse
Affiliation(s)
- Maree L Gould
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Nerida J Downes
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Adele G Woolley
- Maurice Wilkins Centre for Biodiscovery, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Haizal M Hussaini
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
- Faculty of Dental Medicine, University of Airlangga, Surabaya, 60132, Indonesia
| | - Jithendra T Ratnayake
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Mohammad Azam Ali
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Lara T Friedlander
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Paul R Cooper
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| |
Collapse
|
5
|
Murphy JF, Lavelle M, Asciak L, Burdis R, Levis HJ, Ligorio C, McGuire J, Polleres M, Smith PO, Tullie L, Uribe-Gomez J, Chen B, Dawson JI, Gautrot JE, Hooper NM, Kelly DJ, Li VSW, Mata A, Pandit A, Phillips JB, Shu W, Stevens MM, Williams RL, Armstrong JPK, Huang YYS. Biofabrication and biomanufacturing in Ireland and the UK. Biodes Manuf 2024; 7:825-856. [PMID: 39650072 PMCID: PMC11618173 DOI: 10.1007/s42242-024-00316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/11/2024] [Indexed: 12/11/2024]
Abstract
As we navigate the transition from the Fourth to the Fifth Industrial Revolution, the emerging fields of biomanufacturing and biofabrication are transforming life sciences and healthcare. These sectors are benefiting from a synergy of synthetic and engineering biology, sustainable manufacturing, and integrated design principles. Advanced techniques such as 3D bioprinting, tissue engineering, directed assembly, and self-assembly are instrumental in creating biomimetic scaffolds, tissues, organoids, medical devices, and biohybrid systems. The field of biofabrication in the United Kingdom and Ireland is emerging as a pivotal force in bioscience and healthcare, propelled by cutting-edge research and development. Concentrating on the production of biologically functional products for use in drug delivery, in vitro models, and tissue engineering, research institutions across these regions are dedicated to innovating healthcare solutions that adhere to ethical standards while prioritising sustainability, affordability, and healthcare system benefits. Graphic abstract
Collapse
Affiliation(s)
- Jack F. Murphy
- Department of Engineering, University of Cambridge, Cambridge, CB2 1PZ UK
| | - Martha Lavelle
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY UK
| | - Lisa Asciak
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, G4 0NW UK
| | - Ross Burdis
- Department of Materials, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
- Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Hannah J. Levis
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX UK
| | - Cosimo Ligorio
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, NG7 2RD UK
| | - Jamie McGuire
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, S016 6YD UK
| | - Marlene Polleres
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT UK
| | - Poppy O. Smith
- UCL Centre for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX UK
| | - Lucinda Tullie
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, NW1 1AT UK
| | - Juan Uribe-Gomez
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, H91 W2TY Ireland
| | - Biqiong Chen
- School of Mechanical and Aerospace Engineering, Queen’s University Belfast, Belfast, BT9 5AH UK
| | - Jonathan I. Dawson
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, S016 6YD UK
| | - Julien E. Gautrot
- School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS UK
| | - Nigel M. Hooper
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester, M13 9PL UK
| | - Daniel J. Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 H903 Ireland
| | - Vivian S. W. Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, NW1 1AT UK
| | - Alvaro Mata
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, NG7 2RD UK
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD UK
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, H91 W2TY Ireland
| | - James B. Phillips
- UCL Centre for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX UK
| | - Wenmiao Shu
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, G4 0NW UK
| | - Molly M. Stevens
- Department of Materials, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
- Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
- Department of Physiology, Anatomy and Genetics, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU UK
- Department of Engineering Science, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU UK
| | - Rachel L. Williams
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX UK
| | - James P. K. Armstrong
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY UK
| | | |
Collapse
|
6
|
Alaghawani NA, Alkhatib H, Elmancy L, Daou A. Harmonizing Innovations: An In-Depth Comparative Review on the Formulation, Applications, and Future Perspectives of Aerogels and Hydrogels in Pharmaceutical Sciences. Gels 2024; 10:663. [PMID: 39451316 PMCID: PMC11507152 DOI: 10.3390/gels10100663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/21/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024] Open
Abstract
Gels, specifically hydrogels and aerogels, have emerged as versatile materials with profound implications in pharmaceutical sciences. This comprehensive review looks into detail at hydrogels and aerogels, providing a general introduction to gels as a foundation. The paper is then divided into distinct sections for hydrogels and aerogels, each delving into their unique formulations, advantages, disadvantages, and applications. In the realm of hydrogels, we scrutinize the intricacies of formulation, highlighting the versatile advantages they offer. Conversely, potential limitations are explored, paving the way for a detailed discussion on their applications, with a specific focus on their role in antimicrobial applications. Shifting focus to aerogels, a thorough overview is presented, followed by a detailed explanation of the complex formulation process involving sol-gel chemistry; aging; solvent exchange; and drying techniques, including freeze drying, supercritical drying, and ambient-pressure drying (APD). The intricacies of drug loading and release from aerogels are addressed, providing insights into their pharmaceutical potential. The advantages and disadvantages of aerogels are examined, accompanied by an exploration of their applications, with a specific emphasis on antimicrobial uses. The review culminates in a comparative analysis, juxtaposing the advantages and disadvantages of hydrogels and aerogels. Furthermore, the current research and development trends in the applications of these gels in pharmaceutical sciences are discussed, providing a holistic view of their potential and impact. This review serves as a comprehensive guide for researchers, practitioners, and enthusiasts, seeking a deeper understanding of the distinctive attributes and applications of hydrogels and aerogels in the ever-evolving research concerning pharmaceutical sciences.
Collapse
Affiliation(s)
| | | | | | - Anis Daou
- Pharmaceutical Sciences Department, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (N.A.A.); (H.A.); (L.E.)
| |
Collapse
|
7
|
Della Rosa G, Gostynska N, Ephraim JW, Marras S, Moroni M, Tirelli N, Panuccio G, Palazzolo G. Magnesium vs. sodium alginate as precursors of calcium alginate: Mechanical differences and advantages in the development of functional neuronal networks. Carbohydr Polym 2024; 342:122375. [PMID: 39048194 DOI: 10.1016/j.carbpol.2024.122375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 07/27/2024]
Abstract
Calcium alginate is one of the most widely employed matrices in regenerative medicine. A downside is its heterogeneity, due to the poorly controllable character of the gelation of sodium alginate (NaAlg), i.e. the commonly used alginate salt, with calcium. Here, we have used magnesium alginate (MgAlg) as an alternative precursor of calcium alginate. MgAlg coils, more compact and thus less entangled than those of NaAlg, allow for an easier diffusion of calcium ions, whereas Mg is exchanged with calcium more slowly than Na; this allows for the formation of a material (Ca(Mg)Alg) with a more reversible creep behaviour than Ca(Na)Alg, due to a more homogeneous - albeit lower - density of elastically active cross-links. We also show that Ca(Mg)Alg supports better than Ca(Na)Alg the network development and function of embedded (rat cortical) neurons: they show greater neurite extension and branching at 7 and 21 days (Tubb3 and Map2 immunofluorescence) and better neuronal network functional maturation / more robust and longer-lasting activity, probed by calcium imaging and microelectrode array electrophysiology. Overall, our results unveil the potential of MgAlg as bioactive biomaterial for enabling the formation of functional neuron-based tissue analogues.
Collapse
Affiliation(s)
- Giulia Della Rosa
- Istituto Italiano di Tecnologia, Laboratory for Enhanced Regenerative Medicine, Genova, Italy; University of Pavia, Department of Molecular Medicine, Pavia, Italy.
| | - Natalia Gostynska
- Istituto Italiano di Tecnologia, Laboratory for Enhanced Regenerative Medicine, Genova, Italy.
| | - John W Ephraim
- Istituto Italiano di Tecnologia, Laboratory for Enhanced Regenerative Medicine, Genova, Italy.
| | - Sergio Marras
- Istituto Italiano di Tecnologia, Materials Characterization Facility, Genova, Italy.
| | | | - Nicola Tirelli
- Istituto Italiano di Tecnologia, Laboratory for Polymers and Biomaterials, Genova, Italy.
| | - Gabriella Panuccio
- Istituto Italiano di Tecnologia, Laboratory for Enhanced Regenerative Medicine, Genova, Italy.
| | - Gemma Palazzolo
- Istituto Italiano di Tecnologia, Laboratory for Enhanced Regenerative Medicine, Genova, Italy.
| |
Collapse
|
8
|
Yue C, Ding C, Xu M, Hu M, Zhang R. Self-Assembly Behavior of Collagen and Its Composite Materials: Preparation, Characterizations, and Biomedical Engineering and Allied Applications. Gels 2024; 10:642. [PMID: 39451295 PMCID: PMC11507467 DOI: 10.3390/gels10100642] [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: 09/03/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
Collagen is the oldest and most abundant extracellular matrix protein and has many applications in biomedical, food, cosmetic, and other industries. Previous reviews have already introduced collagen's sources, structures, and biosynthesis. The biological and mechanical properties of collagen-based composite materials, their modification and application forms, and their interactions with host tissues are pinpointed. It is worth noting that self-assembly behavior is the main characteristic of collagen molecules. However, there is currently relatively little review on collagen-based composite materials based on self-assembly. Herein, we briefly reviewed the biosynthesis, extraction, structure, and properties of collagen, systematically presented an overview of the various factors and corresponding characterization techniques that affect the collagen self-assembly process, and summarize and discuss the preparation methods and application progress of collagen-based composite materials in different fields. By combining the self-assembly behavior of collagen with preparation methods of collagen-based composite materials, collagen-based composite materials with various functional reactions can be selectively prepared, and these experiences and outcomes can provide inspiration and practical techniques for the future development directions and challenges of collagen-based composite biomaterials in related applications fields.
Collapse
Affiliation(s)
- Chengfei Yue
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (C.Y.)
- Tianjin Key Laboratory of Advanced Fibers and Energy Storage, School of Materials Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Changkun Ding
- Tianjin Key Laboratory of Advanced Fibers and Energy Storage, School of Materials Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Minjie Xu
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (C.Y.)
| | - Min Hu
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (C.Y.)
| | - Ruquan Zhang
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (C.Y.)
| |
Collapse
|
9
|
Niu X, Zhang Y, Wang Y. Co-culture models for investigating cellular crosstalk in the glioma microenvironment. CANCER PATHOGENESIS AND THERAPY 2024; 2:219-230. [PMID: 39371093 PMCID: PMC11447344 DOI: 10.1016/j.cpt.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 10/08/2024]
Abstract
Glioma is the most prevalent primary malignant tumor in the central nervous system (CNS). It represents a diverse group of brain malignancies characterized by the presence of various cancer cell types as well as an array of noncancerous cells, which together form the intricate glioma tumor microenvironment (TME). Understanding the interactions between glioma cells/glioma stem cells (GSCs) and these noncancerous cells is crucial for exploring the pathogenesis and development of glioma. To invesigate these interactions requires in vitro co-culture models that closely mirror the actual TME in vivo. In this review, we summarize the two- and three-dimensional in vitro co-culture model systems for glioma-TME interactions currently available. Furthermore, we explore common glioma-TME cell interactions based on these models, including interactions of glioma cells/GSCs with endothelial cells/pericytes, microglia/macrophages, T cells, astrocytes, neurons, or other multi-cellular interactions. Together, this review provides an update on the glioma-TME interactions, offering insights into glioma pathogenesis.
Collapse
Affiliation(s)
- Xiaodong Niu
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
10
|
Xiao C, Xie N, Shu Q, Liang X, Wang Z, Wu J, Shi N, Huang X, Wei ZC, Gao X, Liu H, Wu K, Xu J, Wang JH, Liu N, Xu F. Synergistic Effects of Matrix Biophysical Properties on Gastric Cancer Cell Behavior via Integrin-Mediated Cell-ECM Interactions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309907. [PMID: 38712486 DOI: 10.1002/smll.202309907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/26/2024] [Indexed: 05/08/2024]
Abstract
The biophysical properties of the extracellular matrix (ECM) play a pivotal role in modulating cancer progression via cell-ECM interactions. However, the biophysical properties specific to gastric cancer (GC) remain largely unexplored. Pertinently, GC ECM shows significantly heterogeneous metamorphoses, such as matrix stiffening and intricate restructuring. By combining collagen I and alginate, this study designs an in vitro biomimetic hydrogel platform to independently modulate matrix stiffness and structure across a physiological stiffness spectrum while preserving consistent collagen concentration and fiber topography. With this platform, this study assesses the impacts of matrix biophysical properties on cell proliferation, migration, invasion, and other pivotal dynamics of AGS. The findings spotlight a compelling interplay between matrix stiffness and structure, influencing both cellular responses and ECM remodeling. Furthermore, this investigation into the integrin/actin-collagen interplay reinforces the central role of integrins in mediating cell-ECM interactions, reciprocally sculpting cell conduct, and ECM adaptation. Collectively, this study reveals a previously unidentified role of ECM biophysical properties in GC malignant potential and provides insight into the bidirectional mechanical cell-ECM interactions, which may facilitate the development of novel therapeutic horizons.
Collapse
Affiliation(s)
- Cailan Xiao
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Ning Xie
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Qiuai Shu
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Xiru Liang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Ziwei Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Jian Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, Shaanxi, 710032, P. R. China
| | - Nianyuan Shi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Key Laboratory of Magnetic Medicine, Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xindi Huang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Zhong-Cao Wei
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Xiaoliang Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, Shaanxi, 710032, P. R. China
| | - Hao Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, Shaanxi, 710032, P. R. China
| | - Kaichun Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, Shaanxi, 710032, P. R. China
| | - Jingyuan Xu
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Department of Gastroenterology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, P. R. China
| | - Jin-Hai Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Na Liu
- Department of Gastroenterology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| |
Collapse
|
11
|
Callegari F, Brofiga M, Tedesco M, Massobrio P. Electrophysiological features of cortical 3D networks are deeply modulated by scaffold properties. APL Bioeng 2024; 8:036112. [PMID: 39193551 PMCID: PMC11348497 DOI: 10.1063/5.0214745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024] Open
Abstract
Three-dimensionality (3D) was proven essential for developing reliable models for different anatomical compartments and many diseases. However, the neuronal compartment still poses a great challenge as we still do not understand precisely how the brain computes information and how the complex chain of neuronal events can generate conscious behavior. Therefore, a comprehensive model of neuronal tissue has not yet been found. The present work was conceived in this framework: we aimed to contribute to what must be a collective effort by filling in some information on possible 3D strategies to pursue. We compared directly different kinds of scaffolds (i.e., PDMS sponges, thermally crosslinked hydrogels, and glass microbeads) in their effect on neuronal network activity recorded using micro-electrode arrays. While the overall rate of spiking activity remained consistent, the type of scaffold had a notable impact on bursting dynamics. The frequency, density of bursts, and occurrence of random spikes were all affected. The examination of inter-burst intervals revealed distinct burst generation patterns unique to different scaffold types. Network burst propagation unveiled divergent trends among configurations. Notably, it showed the most differences, underlying that functional variations may arise from a different 3D spatial organization. This evidence suggests that not all 3D neuronal constructs can sustain the same level of richness of activity. Furthermore, we commented on the reproducibility, efficacy, and scalability of the methods, where the beads still offer superior performances. By comparing different 3D scaffolds, our results move toward understanding the best strategies to develop functional 3D neuronal units for reliable pre-clinical studies.
Collapse
Affiliation(s)
- Francesca Callegari
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
| | | | - Mariateresa Tedesco
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
| | | |
Collapse
|
12
|
Arndt T, Chatterjee U, Shilkova O, Francis J, Lundkvist J, Johansson D, Schmuck B, Greco G, Nordberg ÅE, Li Y, Wahlberg LU, Langton M, Johansson J, Götherström C, Rising A. Tuneable Recombinant Spider Silk Protein Hydrogels for Drug Release and 3D Cell Culture. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2303622. [PMID: 39355087 PMCID: PMC11440629 DOI: 10.1002/adfm.202303622] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/10/2023] [Indexed: 10/03/2024]
Abstract
Hydrogels are useful drug release systems and tissue engineering scaffolds. However, synthetic hydrogels often require harsh gelation conditions and can contain toxic by-products while naturally derived hydrogels can transmit pathogens and in general have poor mechanical properties. Thus, there is a need for a hydrogel that forms under ambient conditions, is non-toxic, xeno-free, and has good mechanical properties. A recombinant spider silk protein-derived hydrogel that rapidly forms at 37 °C is recently developed. The temperature and gelation times are well-suited for an injectable in situ polymerising hydrogel, as well as a 3D cell culture scaffold. Here, it is shown that the diffusion rate and the mechanical properties can be tuned by changing the protein concentration and that human fetal mesenchymal stem cells encapsulated in the hydrogels show high survival and viability. Furthermore, mixtures of recombinant spider silk proteins and green fluorescent protein (GFP) form gels from which functional GFP is gradually released, indicating that bioactive molecules are easily included in the gels, maintain activity and can diffuse through the gel. Interestingly, encapsulated ARPE-19 cells are viable and continuously produce the growth factor progranulin, which is detected in the cell culture medium over the study period of 31 days.
Collapse
Affiliation(s)
- Tina Arndt
- Department of Biosciences and NutritionKarolinska InstitutetNeoHuddinge14152Sweden
| | - Urmimala Chatterjee
- Department of Biosciences and NutritionKarolinska InstitutetNeoHuddinge14152Sweden
| | - Olga Shilkova
- Department of Biosciences and NutritionKarolinska InstitutetNeoHuddinge14152Sweden
| | - Juanita Francis
- Department of Biosciences and NutritionKarolinska InstitutetNeoHuddinge14152Sweden
| | | | - Daniel Johansson
- Department of Molecular SciencesSwedish University of Agricultural SciencesUppsala75007Sweden
| | - Benjamin Schmuck
- Department of Biosciences and NutritionKarolinska InstitutetNeoHuddinge14152Sweden
- Department of AnatomyPhysiology and BiochemistrySwedish University of Agricultural SciencesUppsala75007Sweden
| | - Gabriele Greco
- Department of AnatomyPhysiology and BiochemistrySwedish University of Agricultural SciencesUppsala75007Sweden
| | - Åsa Ekblad Nordberg
- Department of Clinical ScienceIntervention and TechnologyDivision of Obstetrics and GynecologyKarolinska InstitutetHuddinge14152Sweden
| | - Yan Li
- Department of Clinical ScienceIntervention and TechnologyDivision of Orthopedics and BiotechnologyKarolinska UniversitetssjukhusetHuddinge141 86Sweden
| | | | - Maud Langton
- Department of Molecular SciencesSwedish University of Agricultural SciencesUppsala75007Sweden
| | - Jan Johansson
- Department of Biosciences and NutritionKarolinska InstitutetNeoHuddinge14152Sweden
| | - Cecilia Götherström
- Department of Clinical ScienceIntervention and TechnologyDivision of Obstetrics and GynecologyKarolinska InstitutetHuddinge14152Sweden
| | - Anna Rising
- Department of Biosciences and NutritionKarolinska InstitutetNeoHuddinge14152Sweden
- Department of AnatomyPhysiology and BiochemistrySwedish University of Agricultural SciencesUppsala75007Sweden
| |
Collapse
|
13
|
Krsek A, Jagodic A, Baticic L. Nanomedicine in Neuroprotection, Neuroregeneration, and Blood-Brain Barrier Modulation: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1384. [PMID: 39336425 PMCID: PMC11433843 DOI: 10.3390/medicina60091384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024]
Abstract
Nanomedicine is a newer, promising approach to promote neuroprotection, neuroregeneration, and modulation of the blood-brain barrier. This review includes the integration of various nanomaterials in neurological disorders. In addition, gelatin-based hydrogels, which have huge potential due to biocompatibility, maintenance of porosity, and enhanced neural process outgrowth, are reviewed. Chemical modification of these hydrogels, especially with guanidine moieties, has shown improved neuron viability and underscores tailored biomaterial design in neural applications. This review further discusses strategies to modulate the blood-brain barrier-a factor critically associated with the effective delivery of drugs to the central nervous system. These advances bring supportive solutions to the solving of neurological conditions and innovative therapies for their treatment. Nanomedicine, as applied to neuroscience, presents a significant leap forward in new therapeutic strategies that might help raise the treatment and management of neurological disorders to much better levels. Our aim was to summarize the current state-of-knowledge in this field.
Collapse
Affiliation(s)
- Antea Krsek
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
| | - Ana Jagodic
- Department of Family Medicine, Community Health Center Krapina, 49000 Krapina, Croatia;
| | - Lara Baticic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
14
|
Steiner K, Humpel C. Brain Slice Derived Nerve Fibers Grow along Microcontact Prints and are Stimulated by Beta-Amyloid(42). FRONT BIOSCI-LANDMRK 2024; 29:232. [PMID: 38940051 DOI: 10.31083/j.fbl2906232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Alzheimer's disease is characterized by extracellular beta-amyloid plaques, intraneuronal tau neurofibrillary tangles and excessive neurodegeneration. The mechanisms of neuron degeneration and the potential of these neurons to form new nerve fibers for compensation remain elusive. The present study aimed to evaluate the impact of beta-amyloid and tau on new formations of nerve fibers from mouse organotypic brain slices connected to collagen-based microcontact prints. METHODS Organotypic brain slices of postnatal day 8-10 wild-type mice were connected to established collagen-based microcontact prints loaded with polyornithine to enhance nerve fiber outgrowth. Human beta-amyloid(42) or P301S mutated aggregated tau was co-loaded to the prints. Nerve fibers were immunohistochemically stained with neurofilament antibodies. The physiological activity of outgrown neurites was tested with neurotracer MiniRuby, voltage-sensitive dye FluoVolt, and calcium-sensitive dye Rhod-4. RESULTS Immunohistochemical staining revealed newly formed nerve fibers extending along the prints derived from the brain slices. While collagen-only microcontact prints stimulated nerve fiber growth, those loaded with polyornithine significantly enhanced nerve fiber outgrowth. Beta-amyloid(42) significantly increased the neurofilament-positive nerve fibers, while tau had only a weak effect. MiniRuby crystals, retrogradely transported along these newly formed nerve fibers, reached the hippocampus, while FluoVolt and Rhod-4 monitored electrical activity in newly formed nerve fibers. CONCLUSIONS Our data provide evidence that intact nerve fibers can form along collagen-based microcontact prints from mouse brain slices. The Alzheimer's peptide beta-amyloid(42) stimulates this growth, hinting at a neuroprotective function when physiologically active. This "brain-on-chip" model may offer a platform for screening bioactive factors or testing drug effects on nerve fiber growth.
Collapse
Affiliation(s)
- Katharina Steiner
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
15
|
Du G, Zhang J, Shuai Q, Li L, Zhang Q, Shi R. Development of alginate-collagen interpenetrating network for osteoarthritic cartilage by in situ softening. Int J Biol Macromol 2024; 266:131259. [PMID: 38574937 DOI: 10.1016/j.ijbiomac.2024.131259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
This study presents an alginate-collagen interpenetrating network (IPN) matrix of incorporating collagen fibrils into an alginate hydrogel by physical mixing and controlled gelation. The resulting matrix closely mimics the physiological and pathological stiffness range of the chondrocyte pericellular matrix (PCM). Chondrocytes were cultured within three-dimensional (3D) alginate-collagen IPN matrices with varying stiffness, namely Firm, Medium, and Soft. Alginate lyase was introduced to study the effects of the changes in stiffness of the Firm on chondrocyte response by in situ softening. The developed alginate-collagen IPN matrix displayed good cell-biocompatibility. Compared with stiffer tissue culture plastic (TCP), chondrocytes grown within Firm displayed a stabilized differentiated phenotype characterized by higher expression levels of aggrecan, collagen II, and SOX-9. Moreover, the developed alginate-collagen IPN matrix exhibited a gradually increased percentage of propidium iodide (PI)-positive dead cells with decreasing stiffness. Softer matrices directed cells towards higher proliferation rates and spherical morphologies while stimulating chondrocyte cluster formation. Furthermore, reducing Firm stiffness by in situ softening decreased aggrecan expression, contributing to matrix degradation similar to that seen in osteoarthritis (OA). Hence, the 3D alginate-collagen IPN constructs hold significant potential for in vitro replicating PCM stiffness changes observed in OA cartilage.
Collapse
Affiliation(s)
- Genlai Du
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Taiyuan 030001, China.
| | - Jiaqi Zhang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Taiyuan 030001, China
| | - Qizhi Shuai
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Taiyuan 030001, China
| | - Li Li
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Taiyuan 030001, China
| | - Quanyou Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; Department of Orthopaedics, the Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Ruyi Shi
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Taiyuan 030001, China.
| |
Collapse
|
16
|
Kavand A, Noverraz F, Gerber-Lemaire S. Recent Advances in Alginate-Based Hydrogels for Cell Transplantation Applications. Pharmaceutics 2024; 16:469. [PMID: 38675129 PMCID: PMC11053880 DOI: 10.3390/pharmaceutics16040469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
With its exceptional biocompatibility, alginate emerged as a highly promising biomaterial for a large range of applications in regenerative medicine. Whether in the form of microparticles, injectable hydrogels, rigid scaffolds, or bioinks, alginate provides a versatile platform for encapsulating cells and fostering an optimal environment to enhance cell viability. This review aims to highlight recent studies utilizing alginate in diverse formulations for cell transplantation, offering insights into its efficacy in treating various diseases and injuries within the field of regenerative medicine.
Collapse
Affiliation(s)
| | | | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; (A.K.); (F.N.)
| |
Collapse
|
17
|
Yang L, Ni Y, Jiang C, Liu L, Zhang S, Liu J, Sun L, Xu W. A neuromorphic device mimicking synaptic plasticity under different body fluid K + homeostasis for artificial reflex path construction and pattern recognition. FUNDAMENTAL RESEARCH 2024; 4:353-361. [PMID: 38933504 PMCID: PMC11197765 DOI: 10.1016/j.fmre.2022.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022] Open
Abstract
The ionic environment of body fluids influences nervous functions for maintaining homeostasis in organisms and ensures normal perceptual abilities and reflex activities. Neural reflex activities, such as limb movements, are closely associated with potassium ions (K+). In this study, we developed artificial synaptic devices based on ion concentration-adjustable gels for emulating various synaptic plasticities under different K+ concentrations in body fluids. In addition to performing essential synaptic functions, potential applications in information processing and associative learning using short- and long-term plasticity realized using ion concentration-adjustable gels are presented. Artificial synaptic devices can be used for constructing an artificial neural pathway that controls artificial muscle reflex activities and can be used for image pattern recognition. All tests show a strong relationship with ion homeostasis. These devices could be applied to neuromorphic robots and human-machine interfaces.
Collapse
Affiliation(s)
- Lu Yang
- Institute of Photoelectronic Thin Film Devices and Technology, Key Laboratory of Optoelectronic Thin Film Devices and Technology of Tianjin, Engineering Research Center of Thin Film Photoelectronic Technology, Ministry of Education, Nankai University, Tianjin 300350, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518000, China
| | - Yao Ni
- Institute of Photoelectronic Thin Film Devices and Technology, Key Laboratory of Optoelectronic Thin Film Devices and Technology of Tianjin, Engineering Research Center of Thin Film Photoelectronic Technology, Ministry of Education, Nankai University, Tianjin 300350, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518000, China
| | - Chengpeng Jiang
- Institute of Photoelectronic Thin Film Devices and Technology, Key Laboratory of Optoelectronic Thin Film Devices and Technology of Tianjin, Engineering Research Center of Thin Film Photoelectronic Technology, Ministry of Education, Nankai University, Tianjin 300350, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518000, China
| | - Lu Liu
- Institute of Photoelectronic Thin Film Devices and Technology, Key Laboratory of Optoelectronic Thin Film Devices and Technology of Tianjin, Engineering Research Center of Thin Film Photoelectronic Technology, Ministry of Education, Nankai University, Tianjin 300350, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518000, China
| | - Song Zhang
- Institute of Photoelectronic Thin Film Devices and Technology, Key Laboratory of Optoelectronic Thin Film Devices and Technology of Tianjin, Engineering Research Center of Thin Film Photoelectronic Technology, Ministry of Education, Nankai University, Tianjin 300350, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518000, China
| | - Jiaqi Liu
- Institute of Photoelectronic Thin Film Devices and Technology, Key Laboratory of Optoelectronic Thin Film Devices and Technology of Tianjin, Engineering Research Center of Thin Film Photoelectronic Technology, Ministry of Education, Nankai University, Tianjin 300350, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518000, China
| | - Lin Sun
- Institute of Photoelectronic Thin Film Devices and Technology, Key Laboratory of Optoelectronic Thin Film Devices and Technology of Tianjin, Engineering Research Center of Thin Film Photoelectronic Technology, Ministry of Education, Nankai University, Tianjin 300350, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518000, China
| | - Wentao Xu
- Institute of Photoelectronic Thin Film Devices and Technology, Key Laboratory of Optoelectronic Thin Film Devices and Technology of Tianjin, Engineering Research Center of Thin Film Photoelectronic Technology, Ministry of Education, Nankai University, Tianjin 300350, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518000, China
| |
Collapse
|
18
|
Li H, Kuhn M, Kelly RA, Singh A, Palanivel KK, Salama I, De Ieso ML, Stamer WD, Ganapathy PS, Herberg S. Targeting YAP/TAZ mechanosignaling to ameliorate stiffness-induced Schlemm's canal cell pathobiology. Am J Physiol Cell Physiol 2024; 326:C513-C528. [PMID: 38105758 PMCID: PMC11192480 DOI: 10.1152/ajpcell.00438.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
Pathological alterations in the biomechanical properties of the Schlemm's canal (SC) inner wall endothelium and its immediate vicinity are strongly associated with ocular hypertension in glaucoma due to decreased outflow facility. Specifically, the underlying trabecular meshwork is substantially stiffer in glaucomatous eyes compared with that from normal eyes. This raises the possibility of a critical involvement of mechanotransduction processes in driving SC cell dysfunction. Yes-associated protein (YAP) has emerged as a key contributor to glaucoma pathogenesis. However, the molecular underpinnings of SC cell mechanosignaling via YAP and transcriptional coactivator with PDZ-binding motif (TAZ) in response to glaucomatous extracellular matrix (ECM) stiffening are not well understood. Using a novel biopolymer hydrogel that facilitates dynamic and reversible stiffness tuning, we investigated how ECM stiffening modulates YAP/TAZ activity in primary human SC cells, and whether disruption of YAP/TAZ mechanosignaling attenuates SC cell pathobiology and increases ex vivo outflow facility. We demonstrated that ECM stiffening drives pathologic YAP/TAZ activation and cytoskeletal reorganization in SC cells, which was fully reversible by matrix softening in a distinct time-dependent manner. Furthermore, we showed that pharmacologic or genetic disruption of YAP/TAZ mechanosignaling abrogates stiffness-induced SC cell dysfunction involving altered cytoskeletal and ECM remodeling. Finally, we found that perfusion of the clinically used, small molecule YAP/TAZ inhibitor verteporfin (without light activation) increases ex vivo outflow facility in normal mouse eyes. Collectively, our data provide new evidence for a pathologic role of aberrant YAP/TAZ mechanosignaling in SC cell dysfunction and suggest that YAP/TAZ inhibition has therapeutic value for treating ocular hypertension in glaucoma.NEW & NOTEWORTHY Pathologically altered biomechanical properties of the Schlemm's canal (SC) inner wall microenvironment were recently validated as the cause for increased outflow resistance in ocular hypertensive glaucoma. However, the involvement of specific mechanotransduction pathways in these disease processes is largely unclear. Here, we demonstrate that Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) are central regulators of glaucoma-like SC cell dysfunction in response to extracellular matrix stiffening and that targeted disruption of YAP/TAZ mechanosignaling attenuates SC cell pathobiology and enhances outflow function.
Collapse
Affiliation(s)
- Haiyan Li
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, United States
- BioInspired Institute, Syracuse University, Syracuse, New York, United States
| | - Megan Kuhn
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, North Carolina, United States
| | - Ruth A Kelly
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, North Carolina, United States
| | - Ayushi Singh
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, United States
- BioInspired Institute, Syracuse University, Syracuse, New York, United States
| | - Kavipriya Kovai Palanivel
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
| | - Izzy Salama
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
| | - Michael L De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, North Carolina, United States
| | - W Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, North Carolina, United States
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
| | - Preethi S Ganapathy
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
- BioInspired Institute, Syracuse University, Syracuse, New York, United States
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, United States
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, United States
- BioInspired Institute, Syracuse University, Syracuse, New York, United States
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, United States
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York, United States
| |
Collapse
|
19
|
Desai SU, Srinivasan SS, Kumbar SG, Moss IL. Hydrogel-Based Strategies for Intervertebral Disc Regeneration: Advances, Challenges and Clinical Prospects. Gels 2024; 10:62. [PMID: 38247785 PMCID: PMC10815657 DOI: 10.3390/gels10010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Millions of people worldwide suffer from low back pain and disability associated with intervertebral disc (IVD) degeneration. IVD degeneration is highly correlated with aging, as the nucleus pulposus (NP) dehydrates and the annulus fibrosus (AF) fissures form, which often results in intervertebral disc herniation or disc space collapse and related clinical symptoms. Currently available options for treating intervertebral disc degeneration are symptoms control with therapy modalities, and/or medication, and/or surgical resection of the IVD with or without spinal fusion. As such, there is an urgent clinical demand for more effective disease-modifying treatments for this ubiquitous disorder, rather than the current paradigms focused only on symptom control. Hydrogels are unique biomaterials that have a variety of distinctive qualities, including (but not limited to) biocompatibility, highly adjustable mechanical characteristics, and most importantly, the capacity to absorb and retain water in a manner like that of native human nucleus pulposus tissue. In recent years, various hydrogels have been investigated in vitro and in vivo for the repair of intervertebral discs, some of which are ready for clinical testing. In this review, we summarize the latest findings and developments in the application of hydrogel technology for the repair and regeneration of intervertebral discs.
Collapse
Affiliation(s)
- Shivam U. Desai
- Department of Orthopedic Surgery, Central Michigan University, College of Medicine, Saginaw, MI 48602, USA
| | | | | | - Isaac L. Moss
- Department of Orthopedic Surgery, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
20
|
Pereira I, Lopez-Martinez MJ, Samitier J. Advances in current in vitro models on neurodegenerative diseases. Front Bioeng Biotechnol 2023; 11:1260397. [PMID: 38026882 PMCID: PMC10658011 DOI: 10.3389/fbioe.2023.1260397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Many neurodegenerative diseases are identified but their causes and cure are far from being well-known. The problem resides in the complexity of the neural tissue and its location which hinders its easy evaluation. Although necessary in the drug discovery process, in vivo animal models need to be reduced and show relevant differences with the human tissues that guide scientists to inquire about other possible options which lead to in vitro models being explored. From organoids to organ-on-a-chips, 3D models are considered the cutting-edge technology in cell culture. Cell choice is a big parameter to take into consideration when planning an in vitro model and cells capable of mimicking both healthy and diseased tissue, such as induced pluripotent stem cells (iPSC), are recognized as good candidates. Hence, we present a critical review of the latest models used to study neurodegenerative disease, how these models have evolved introducing microfluidics platforms, 3D cell cultures, and the use of induced pluripotent cells to better mimic the neural tissue environment in pathological conditions.
Collapse
Affiliation(s)
- Inês Pereira
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria J. Lopez-Martinez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Read SA, Go CS, Ferreira MJS, Ligorio C, Kimber SJ, Dumanli AG, Domingos MAN. Nanocrystalline Cellulose as a Versatile Engineering Material for Extrusion-Based Bioprinting. Pharmaceutics 2023; 15:2432. [PMID: 37896192 PMCID: PMC10609932 DOI: 10.3390/pharmaceutics15102432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Naturally derived polysaccharide-based hydrogels, such as alginate, are frequently used in the design of bioinks for 3D bioprinting. Traditionally, the formulation of such bioinks requires the use of pre-reticulated materials with low viscosities, which favour cell viability but can negatively influence the resolution and shape fidelity of the printed constructs. In this work, we propose the use of cellulose nanocrystals (CNCs) as a rheological modifier to improve the printability of alginate-based bioinks whilst ensuring a high viability of encapsulated cells. Through rheological analysis, we demonstrate that the addition of CNCs (1% and 2% (w/v)) to alginate hydrogels (1% (w/v)) improves shear-thinning behaviour and mechanical stability, resulting in the high-fidelity printing of constructs with superior resolution. Importantly, LIVE/DEAD results confirm that the presence of CNCs does not seem to affect the health of immortalised chondrocytes (TC28a2) that remain viable over a period of seven days post-encapsulation. Taken together, our results indicate a favourable effect of the CNCs on the rheological and biocompatibility properties of alginate hydrogels, opening up new perspectives for the application of CNCs in the formulation of bioinks for extrusion-based bioprinting.
Collapse
Affiliation(s)
- Sophia A. Read
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, The University of Manchester, Manchester M13 9PL, UK; (S.A.R.); (C.S.G.); (M.J.S.F.)
| | - Chee Shuen Go
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, The University of Manchester, Manchester M13 9PL, UK; (S.A.R.); (C.S.G.); (M.J.S.F.)
| | - Miguel J. S. Ferreira
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, The University of Manchester, Manchester M13 9PL, UK; (S.A.R.); (C.S.G.); (M.J.S.F.)
| | - Cosimo Ligorio
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering & Henry Royce Institute, The University of Manchester, Manchester M13 9PL, UK; (C.L.); (A.G.D.)
| | - Susan J. Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Ahu G. Dumanli
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering & Henry Royce Institute, The University of Manchester, Manchester M13 9PL, UK; (C.L.); (A.G.D.)
| | - Marco A. N. Domingos
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, The University of Manchester, Manchester M13 9PL, UK; (S.A.R.); (C.S.G.); (M.J.S.F.)
| |
Collapse
|
22
|
Wang S, Bai L, Hu X, Yao S, Hao Z, Zhou J, Li X, Lu H, He J, Wang L, Li D. 3D Bioprinting of Neurovascular Tissue Modeling with Collagen-Based Low-Viscosity Composites. Adv Healthc Mater 2023; 12:e2300004. [PMID: 37264745 PMCID: PMC11469067 DOI: 10.1002/adhm.202300004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 05/27/2023] [Indexed: 06/03/2023]
Abstract
In vitro neurovascular unit (NVU) models are valuable for investigating brain functions and developing drugs. However, it remains challenging to recapitulate the native architectural features and ultra-soft extracellular matrix (ECM) properties of the natural NVU. Cell-laden bioprinting is promising to prepare complex living tissues, but hard to balance the fidelity and cell growth. This study proposes a novel two-stage methodology for biomanufacturing functional 3D neurovascular constructs in vitro with low modulus of ECM. At the shaping stage, a low-viscosity alginate/collagen is printed through an embedded approach; at the culturing stage, the alginate is removed through targeted lysing. The low-viscosity and rapid crosslinking properties provide a printing resolution of ≈10 µm, and the lysis processing can decrease the hydrogels' modulus to ≈1 kPa and adjust the porosity of the microstructure, providing cells with an environment closing to the brain ECM. A 3D hollow coaxial neurovascular model is fabricated, in which the endothelial cells has expressed tight junction proteins and shown selective permeability, and the astrocytes outside of the endothelial layer are found to spread out with branches and directly interact with endothelial cells. The present study offers a promising modeling method for better understanding the NVU function and screening neuro-drugs.
Collapse
Affiliation(s)
- Sen Wang
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Luge Bai
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Xiaoxuan Hu
- Institute of NeurobiologySchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'an710061China
- Key Laboratory of Ministry of Education for Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterXi'an710061China
| | - Siqi Yao
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Zhiyan Hao
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - JiaJia Zhou
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Xiao Li
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Haixia Lu
- Institute of NeurobiologySchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'an710061China
- Key Laboratory of Ministry of Education for Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterXi'an710061China
- Department of Human Anatomy & HistoembryologySchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'an710061China
| | - Jiankang He
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Ling Wang
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| | - Dichen Li
- State Key Laboratory for Manufacturing System EngineeringXi'an Jiaotong UniversityXi'an710054China
- School of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
- NMPA Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an710054China
| |
Collapse
|
23
|
Li H, Kuhn M, Kelly RA, Singh A, Palanivel KK, Salama I, De Ieso ML, Stamer WD, Ganapathy PS, Herberg S. Targeting YAP mechanosignaling to ameliorate stiffness-induced Schlemm's canal cell pathobiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.08.556840. [PMID: 37781615 PMCID: PMC10541092 DOI: 10.1101/2023.09.08.556840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Pathologic alterations in the biomechanical properties of the Schlemm's canal (SC) inner wall endothelium and its immediate vicinity are strongly associated with ocular hypertension in glaucoma due to decreased outflow facility. Specifically, the underlying trabecular meshwork is substantially stiffer in glaucomatous eyes compared to that from normal eyes. This raises the possibility of a critical involvement of mechanotransduction processes in driving SC cell dysfunction. Yes-associated protein (YAP) has emerged as a key contributor to glaucoma pathogenesis. However, the molecular underpinnings of SC cell YAP mechanosignaling in response to glaucomatous extracellular matrix (ECM) stiffening are not well understood. Using a novel biopolymer hydrogel that facilitates dynamic and reversible stiffness tuning, we investigated how ECM stiffening modulates YAP activity in primary human SC cells, and whether disruption of YAP mechanosignaling attenuates SC cell pathobiology and increases ex vivo outflow facility. We demonstrated that ECM stiffening drives pathologic YAP activation and cytoskeletal reorganization in SC cells, which was fully reversible by matrix softening in a distinct time-dependent manner. Furthermore, we showed that pharmacologic or genetic disruption of YAP mechanosignaling abrogates stiffness-induced SC cell dysfunction involving altered cytoskeletal and ECM remodeling. Lastly, we found that perfusion of the clinically-used, small molecule YAP inhibitor verteporfin (without light activation) increases ex vivo outflow facility in normal mouse eyes. Collectively, our data provide new evidence for a pathologic role of aberrant YAP mechanosignaling in SC cell dysfunction and suggest that YAP inhibition has therapeutic value for treating ocular hypertension in glaucoma.
Collapse
Affiliation(s)
- Haiyan Li
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Megan Kuhn
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27708, USA
| | - Ruth A. Kelly
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27708, USA
| | - Ayushi Singh
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Kavipriya Kovai Palanivel
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Izzy Salama
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Michael L. De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27708, USA
| | - W. Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Preethi S. Ganapathy
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
24
|
Lipreri MV, Di Pompo G, Boanini E, Graziani G, Sassoni E, Baldini N, Avnet S. Bone on-a-chip: a 3D dendritic network in a screening platform for osteocyte-targeted drugs. Biofabrication 2023; 15:045019. [PMID: 37552982 DOI: 10.1088/1758-5090/acee23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 08/08/2023] [Indexed: 08/10/2023]
Abstract
Age-related musculoskeletal disorders, including osteoporosis, are frequent and associated with long lasting morbidity, in turn significantly impacting on healthcare system sustainability. There is therefore a compelling need to develop reliable preclinical models of disease and drug screening to validate novel drugs possibly on a personalized basis, without the need ofin vivoassay. In the context of bone tissue, although the osteocyte (Oc) network is a well-recognized therapeutic target, currentin vitropreclinical models are unable to mimic its physiologically relevant and highly complex structure. To this purpose, several features are needed, including an osteomimetic extracellular matrix, dynamic perfusion, and mechanical cues (e.g. shear stress) combined with a three-dimensional (3D) culture of Oc. Here we describe, for the first time, a high throughput microfluidic platform based on 96-miniaturized chips for large-scale preclinical evaluation to predict drug efficacy. We bioengineered a commercial microfluidic device that allows real-time visualization and equipped with multi-chips by the development and injection of a highly stiff bone-like 3D matrix, made of a blend of collagen-enriched natural hydrogels loaded with hydroxyapatite nanocrystals. The microchannel, filled with the ostemimetic matrix and Oc, is subjected to passive perfusion and shear stress. We used scanning electron microscopy for preliminary material characterization. Confocal microscopy and fluorescent microbeads were used after material injection into the microchannels to detect volume changes and the distribution of cell-sized objects within the hydrogel. The formation of a 3D dendritic network of Oc was monitored by measuring cell viability, evaluating phenotyping markers (connexin43, integrin alpha V/CD51, sclerostin), quantification of dendrites, and responsiveness to an anabolic drug. The platform is expected to accelerate the development of new drug aimed at modulating the survival and function of osteocytes.
Collapse
Affiliation(s)
| | - Gemma Di Pompo
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Boanini
- Department of Chemistry 'Giacomo Ciamician', University of Bologna, Bologna, Italy
| | - Gabriela Graziani
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Enrico Sassoni
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, Bologna, Italy
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
25
|
Fuenteslópez CV, Thompson MS, Ye H. Development and Optimisation of Hydrogel Scaffolds for Microvascular Network Formation. Bioengineering (Basel) 2023; 10:964. [PMID: 37627849 PMCID: PMC10451297 DOI: 10.3390/bioengineering10080964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Traumatic injuries are a major cause of morbidity and mortality worldwide; however, there is limited research on microvascular traumatic injuries. To address this gap, this research aims to develop and optimise an in vitro construct for traumatic injury research at the microvascular level. Tissue engineering constructs were created using a range of polymers (collagen, fibrin, and gelatine), solvents (PBS, serum-free endothelial media, and MES/NaCl buffer), and concentrations (1-5% w/v). Constructs created from these hydrogels and HUVECs were evaluated to identify the optimal composition in terms of cell proliferation, adhesion, migration rate, viability, hydrogel consistency and shape retention, and tube formation. Gelatine hydrogels were associated with a lower cell adhesion, whereas fibrin and collagen ones displayed similar or better results than the control, and collagen hydrogels exhibited poor shape retention; fibrin scaffolds, particularly at high concentrations, displayed good hydrogel consistency. Based on the multipronged evaluation, fibrin hydrogels in serum-free media at 3 and 5% w/v were selected for further experimental work and enabled the formation of interconnected capillary-like networks. The networks formed in both hydrogels displayed a similar architecture in terms of the number of segments (10.3 ± 3.21 vs. 9.6 ± 3.51) and diameter (8.6446 ± 3.0792 μm vs. 7.8599 ± 2.3794 μm).
Collapse
Affiliation(s)
| | | | - Hua Ye
- Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK; (C.V.F.); (M.S.T.)
| |
Collapse
|
26
|
Babaei A, Tiraihi T, Ai J, Baheiraei N. Enhanced growth and differentiation of neural stem cells on alginate/collagen/reduced graphene oxide composite hydrogel incorporated with lithium chloride. BIOIMPACTS : BI 2023; 13:475-487. [PMID: 38022379 PMCID: PMC10676529 DOI: 10.34172/bi.2023.24266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 12/01/2023]
Abstract
Introduction Cell transplantation with hydrogel-based carriers is one of the advanced therapeutics for challenging diseases, such as spinal cord injury. Electrically conductive hydrogel has received much attention for its effect on nerve outgrowth and differentiation. Besides, a load of neuroprotective substances, such as lithium chloride can promote the differentiation properties of the hydrogel. Methods In this study, alginate/collagen/reduced graphene oxide hydrogel loaded with lithium chloride (AL/CO/rGO Li+) was prepared as an injectable cell delivery system for neural tissue regeneration. After determining the lithium-ion release profile, an MTT assay was performed to check neural viability. In the next step, real-time PCR was performed to evaluate the expression of cell adhesion and neurogenic markers. Results Our results showed that the combination of collagen fibers and rGO with alginates increased cell viability and the gene expression of collagen-binding receptor subunits such as integrin α1, and β1. Further, rGO contributed to the controlled release of lithium-ion hydrogel in terms of its plenty of negatively charged functional groups. The continuous culture of NSCs on AL/CO/rGO Li+ hydrogel increased neurogenic genes' expressions of nestin (5.9 fold), NF200 (36.8 fold), and synaptophysin (13.2 fold), as well as protein expression of NF200 and synaptophysin after about 14 days. Conclusion The simultaneous ability of electrical conduction and lithium-ion release of AL/CO/rGO Li+ hydrogel could provide a favorable microenvironment for NSCs by improving their survival, maintaining cell morphology, and expressing the neural marker. It may be potentially used as a therapeutic approach for stem cell transplantation in a spinal cord injury.
Collapse
Affiliation(s)
- Azadeh Babaei
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Taki Tiraihi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Jajar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Baheiraei
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
27
|
Kuznetsova YL, Gushchina KS, Lobanova KS, Chasova VO, Egorikhina MN, Grigoreva AO, Malysheva YB, Kuzmina DA, Farafontova EA, Linkova DD, Rubtsova YP, Semenycheva LL. Scaffold Chemical Model Based on Collagen-Methyl Methacrylate Graft Copolymers. Polymers (Basel) 2023; 15:2618. [PMID: 37376264 DOI: 10.3390/polym15122618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Polymerization of methyl methacrylate (MMA) in aqueous collagen (Col) dispersion was studied in the presence of tributylborane (TBB) and p-quinone: 2,5-di-tert-butyl-p-benzoquinone (2,5-DTBQ), p-benzoquinone (BQ), duroquinone (DQ), and p-naphthoquinone (NQ). It was found that this system leads to the formation of a grafted cross-linked copolymer. The inhibitory effect of p-quinone determines the amount of unreacted monomer, homopolymer, and percentage of grafted poly(methyl methacrylate) (PMMA). The synthesis combines two approaches to form a grafted copolymer with a cross-linked structure-"grafting to" and "grafting from". The resulting products exhibit biodegradation under the action of enzymes, do not have toxicity, and demonstrate a stimulating effect on cell growth. At the same time, the denaturation of collagen occurring at elevated temperatures does not impair the characteristics of copolymers. These results allow us to present the research as a scaffold chemical model. Comparison of the properties of the obtained copolymers helps to determine the optimal method for the synthesis of scaffold precursors-synthesis of a collagen and poly(methyl methacrylate) copolymer at 60 °C in a 1% acetic acid dispersion of fish collagen with a mass ratio of the components collagen:MMA:TBB:2,5-DTBQ equal to 1:1:0.015:0.25.
Collapse
Affiliation(s)
- Yulia L Kuznetsova
- Faculty of Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23, Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Ksenya S Gushchina
- Faculty of Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23, Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Karina S Lobanova
- Faculty of Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23, Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Victoria O Chasova
- Faculty of Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23, Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Marfa N Egorikhina
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia
| | - Alexandra O Grigoreva
- Faculty of Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23, Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Yulia B Malysheva
- Faculty of Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23, Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Daria A Kuzmina
- Faculty of Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23, Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Ekaterina A Farafontova
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia
| | - Daria D Linkova
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia
| | - Yulia P Rubtsova
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia
| | - Luydmila L Semenycheva
- Faculty of Chemistry, National Research Lobachevsky State University of Nizhny Novgorod, 23, Gagarin Ave., 603022 Nizhny Novgorod, Russia
| |
Collapse
|
28
|
Huang WH, Ding SL, Zhao XY, Li K, Guo HT, Zhang MZ, Gu Q. Collagen for neural tissue engineering: Materials, strategies, and challenges. Mater Today Bio 2023; 20:100639. [PMID: 37197743 PMCID: PMC10183670 DOI: 10.1016/j.mtbio.2023.100639] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/19/2023] Open
Abstract
Neural tissue engineering (NTE) has made remarkable strides in recent years and holds great promise for treating several devastating neurological disorders. Selecting optimal scaffolding material is crucial for NET design strategies that enable neural and non-neural cell differentiation and axonal growth. Collagen is extensively employed in NTE applications due to the inherent resistance of the nervous system against regeneration, functionalized with neurotrophic factors, antagonists of neural growth inhibitors, and other neural growth-promoting agents. Recent advancements in integrating collagen with manufacturing strategies, such as scaffolding, electrospinning, and 3D bioprinting, provide localized trophic support, guide cell alignment, and protect neural cells from immune activity. This review categorises and analyses collagen-based processing techniques investigated for neural-specific applications, highlighting their strengths and weaknesses in repair, regeneration, and recovery. We also evaluate the potential prospects and challenges of using collagen-based biomaterials in NTE. Overall, this review offers a comprehensive and systematic framework for the rational evaluation and applications of collagen in NTE.
Collapse
Affiliation(s)
- Wen-Hui Huang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
| | - Sheng-Long Ding
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, PR China
| | - Xi-Yuan Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
| | - Kai Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
| | - Hai-Tao Guo
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
| | - Ming-Zhu Zhang
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, PR China
- Corresponding author.
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
- Corresponding author. Institute of Zoology, Chinese Academy of Sciences, No. 5 of Courtyard 1, Beichen West Road, Chaoyang District, Beijing 100101, PR China.
| |
Collapse
|
29
|
Whitehouse C, Corbett N, Brownlees J. 3D models of neurodegeneration: implementation in drug discovery. Trends Pharmacol Sci 2023; 44:208-221. [PMID: 36822950 DOI: 10.1016/j.tips.2023.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/24/2023]
Abstract
A lack of in vitro models that robustly represent the complex cellular pathologies underlying neurodegeneration has resulted in a translational gap between in vitro and in vivo results, creating a bottleneck in the development of new therapeutics. In the past decade, new and complex 3D models of the brain have been published at an exponential rate. However, many novel 3D models of neurodegeneration overlook the validation and throughput requirements for implementation in drug discovery. This therefore represents a knowledge gap that could hinder the translation of these models to drug discovery efforts. We review the recent progress in the development of 3D models of neurodegeneration, examining model design benefits and validation techniques, and discuss opportunities and standards for 3D models of neurodegeneration to be implemented in drug discovery and development.
Collapse
Affiliation(s)
| | - Nicola Corbett
- MSD R&D Innovation Centre, 120 Moorgate, London EC2M 6UR, UK
| | - Janet Brownlees
- MSD R&D Innovation Centre, 120 Moorgate, London EC2M 6UR, UK
| |
Collapse
|
30
|
Wang T, Yu T, Tsai CY, Hong ZY, Chao WH, Su YS, Subbiah SK, Renuka RR, Hsu ST, Wu GJ, Higuchi A. Xeno-free culture and proliferation of hPSCs on 2D biomaterials. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 199:63-107. [PMID: 37678982 DOI: 10.1016/bs.pmbts.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Human pluripotent stem cells (human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs)) have unlimited proliferative potential, whereas adult stem cells such as bone marrow-derived stem cells and adipose-derived stem cells have problems with aging. When hPSCs are intended to be cultured on feeder-free or xeno-free conditions without utilizing mouse embryonic fibroblasts or human fibroblasts, they cannot be cultured on conventional tissue culture polystyrene dishes, as adult stem cells can be cultured but should be cultivated on material surfaces grafted or coated with (a) natural or recombinant extracellular matrix (ECM) proteins, (b) ECM protein-derived peptides and specific synthetic polymer surfaces in xeno-free and/or chemically defined conditions. This review describes current developing cell culture biomaterials for the proliferation of hPSCs while maintaining the pluripotency and differentiation potential of the cells into 3 germ layers. Biomaterials for the cultivation of hPSCs without utilizing a feeder layer are essential to decrease the risk of xenogenic molecules, which contributes to the potential clinical usage of hPSCs. ECM proteins such as human recombinant vitronectin, laminin-511 and laminin-521 have been utilized instead of Matrigel for the feeder-free cultivation of hPSCs. The following biomaterials are also discussed for hPSC cultivation: (a) decellularized ECM, (b) peptide-grafted biomaterials derived from ECM proteins, (c) recombinant E-cadherin-coated surface, (d) polysaccharide-immobilized surface, (e) synthetic polymer surfaces with and without bioactive sites, (f) thermoresponsive polymer surfaces with and without bioactive sites, and (g) synthetic microfibrous scaffolds.
Collapse
Affiliation(s)
- Ting Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Tao Yu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Chang-Yen Tsai
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Zhao-Yu Hong
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Wen-Hui Chao
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Yi-Shuo Su
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| | - Remya Rajan Renuka
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| | - Shih-Tien Hsu
- Department of Internal Medicine, Landseed International Hospital, Pingjen City, Taoyuan, Taiwan
| | - Gwo-Jang Wu
- Graduate Institute of Medical Sciences and Department of Obstetrics & Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Akon Higuchi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Graduate Institute of Medical Sciences and Department of Obstetrics & Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
31
|
Afshar A, Gultekinoglu M, Edirisinghe M. Binary polymer systems for biomedical applications. INTERNATIONAL MATERIALS REVIEWS 2023; 68:184-224. [DOI: 10.1080/09506608.2022.2069451] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 04/08/2022] [Indexed: 01/06/2025]
Affiliation(s)
- Ayda Afshar
- Department of Mechanical Engineering, University College London, London, UK
| | - Merve Gultekinoglu
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Mohan Edirisinghe
- Department of Mechanical Engineering, University College London, London, UK
| |
Collapse
|
32
|
Ma H, Yu K, Wang H, Liu J, Cheng YY, Kang Y, Wang H, Zhang J, Song K. Fabrication and detection of a novel hybrid conductive scaffold based on alginate/gelatin/carboxylated carbon nanotubes (Alg/Gel/mMWCNTs) for neural tissue engineering. Tissue Cell 2023; 80:101995. [PMID: 36512950 DOI: 10.1016/j.tice.2022.101995] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
Carbon nanotubes (CNTs), as kinds of conductive carbon nanomaterials, were widely applied in neural tissue engineering due to their excellent electrical conductivity and good biocompatibility. In this study, the carboxyl-modified multi-walled carbon nanotubes (mMWCNTs) were introduced into sodium alginate/gelatin (Alg/Gel) scaffolds to optimize the function of the hybrid scaffolds. The Alg/Gel/mMWCNTs conductive scaffolds with mMWCNTs content of 1%, 3%, and 5% were prepared by freeze-drying, respectively. Following this, the physicochemical properties and biocompatibility of the hybrid scaffolds at different magnetic field intensities were evaluated. The conductive scaffolds were characterized by Scanning electron microscopy (SEM) and Fourier transform infrared spectroscopy (FTIR). In general, the mMWCNTs addition improved the hydrophilic, electrical conductivity and mechanical properties of the composite scaffold, and PC12 cells showed a trend of gradual increase over culture time. Particularly, the Alg/Gel-1%C scaffold exhibited the best cell proliferation behavior. Briefly, the surface contact angle decreased from 74 ± 1° to 60 ± 3°, the electrical conductivity and compressive modulus increased to 1.32 × 10-3 ± 2.1 × 10-4 S/cm and 1.40 ± 0.076 Mpa, the G1 phase from 55.67 ± 1.86% to 59.77 ± 0.94% and the G2 phase from 10.32 ± 0.35% to 13.93 ± 1.26%,respectively. In the SEM images, PC12 cells were well-shaped and densely distributed. Therefore, the Alg/Gel/mMWCNTs conductive scaffold has potential as a tissue engineering scaffold in nerve regeneration.
Collapse
Affiliation(s)
- Hailin Ma
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Kai Yu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Hao Wang
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Jiaqi Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Yue Kang
- Department of Breast Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China.
| | - Hong Wang
- Department of Orthopeadics, Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian 116033, China.
| | - Jingying Zhang
- The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808 Guangdong, China.
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China.
| |
Collapse
|
33
|
Ma C, Duan X, Lei X. 3D cell culture model: From ground experiment to microgravity study. Front Bioeng Biotechnol 2023; 11:1136583. [PMID: 37034251 PMCID: PMC10080128 DOI: 10.3389/fbioe.2023.1136583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Microgravity has been shown to induce many changes in cell growth and differentiation due to offloading the gravitational strain normally exerted on cells. Although many studies have used two-dimensional (2D) cell culture systems to investigate the effects of microgravity on cell growth, three-dimensional (3D) culture scaffolds can offer more direct indications of the modified cell response to microgravity-related dysregulations compared to 2D culture methods. Thus, knowledge of 3D cell culture is essential for better understanding the in vivo tissue function and physiological response under microgravity conditions. This review discusses the advances in 2D and 3D cell culture studies, particularly emphasizing the role of hydrogels, which can provide cells with a mimic in vivo environment to collect a more natural response. We also summarized recent studies about cell growth and differentiation under real microgravity or simulated microgravity conditions using ground-based equipment. Finally, we anticipate that hydrogel-based 3D culture models will play an essential role in constructing organoids, discovering the causes of microgravity-dependent molecular and cellular changes, improving space tissue regeneration, and developing innovative therapeutic strategies. Future research into the 3D culture in microgravity conditions could lead to valuable therapeutic applications in health and pharmaceuticals.
Collapse
Affiliation(s)
- Chiyuan Ma
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Xianglong Duan
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
- Second Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, China
- *Correspondence: Xianglong Duan, ; Xiaohua Lei,
| | - Xiaohua Lei
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- *Correspondence: Xianglong Duan, ; Xiaohua Lei,
| |
Collapse
|
34
|
Message in a Scaffold: Natural Biomaterials for Three-Dimensional (3D) Bioprinting of Human Brain Organoids. Biomolecules 2022; 13:biom13010025. [PMID: 36671410 PMCID: PMC9855696 DOI: 10.3390/biom13010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Brain organoids are invaluable tools for pathophysiological studies or drug screening, but there are still challenges to overcome in making them more reproducible and relevant. Recent advances in three-dimensional (3D) bioprinting of human neural organoids is an emerging approach that may overcome the limitations of self-organized organoids. It requires the development of optimal hydrogels, and a wealth of research has improved our knowledge about biomaterials both in terms of their intrinsic properties and their relevance on 3D culture of brain cells and tissue. Although biomaterials are rarely biologically neutral, few articles have reviewed their roles on neural cells. We here review the current knowledge on unmodified biomaterials amenable to support 3D bioprinting of neural organoids with a particular interest in their impact on cell homeostasis. Alginate is a particularly suitable bioink base for cell encapsulation. Gelatine is a valuable helper agent for 3D bioprinting due to its viscosity. Collagen, fibrin, hyaluronic acid and laminin provide biological support to adhesion, motility, differentiation or synaptogenesis and optimize the 3D culture of neural cells. Optimization of specialized hydrogels to direct differentiation of stem cells together with an increased resolution in phenotype analysis will further extend the spectrum of possible bioprinted brain disease models.
Collapse
|
35
|
Liu R, Meng X, Yu X, Wang G, Dong Z, Zhou Z, Qi M, Yu X, Ji T, Wang F. From 2D to 3D Co-Culture Systems: A Review of Co-Culture Models to Study the Neural Cells Interaction. Int J Mol Sci 2022; 23:13116. [PMID: 36361902 PMCID: PMC9656609 DOI: 10.3390/ijms232113116] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 06/11/2024] Open
Abstract
The central nervous system (CNS) controls and regulates the functional activities of the organ systems and maintains the unity between the body and the external environment. The advent of co-culture systems has made it possible to elucidate the interactions between neural cells in vitro and to reproduce complex neural circuits. Here, we classified the co-culture system as a two-dimensional (2D) co-culture system, a cell-based three-dimensional (3D) co-culture system, a tissue slice-based 3D co-culture system, an organoid-based 3D co-culture system, and a microfluidic platform-based 3D co-culture system. We provide an overview of these different co-culture models and their applications in the study of neural cell interaction. The application of co-culture systems in virus-infected CNS disease models is also discussed here. Finally, the direction of the co-culture system in future research is prospected.
Collapse
Affiliation(s)
- Rongrong Liu
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xiaoting Meng
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xiyao Yu
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Guoqiang Wang
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Zhiyong Dong
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Zhengjie Zhou
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Mingran Qi
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xiao Yu
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Tong Ji
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fang Wang
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
36
|
Sodium Alginate—Natural Microencapsulation Material of Polymeric Microparticles. Int J Mol Sci 2022; 23:ijms232012108. [PMID: 36292962 PMCID: PMC9603258 DOI: 10.3390/ijms232012108] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/30/2022] Open
Abstract
From the multitude of materials currently available on the market that can be used in the development of microparticles, sodium alginate has become one of the most studied natural anionic polymers that can be included in controlled-release pharmaceutical systems alongside other polymers due to its low cost, low toxicity, biocompatibility, biodegradability and gelatinous die-forming capacity in the presence of Ca2+ ions. In this review, we have shown that through coacervation, the particulate systems for the dispensing of drugs consisting of natural polymers are nontoxic, allowing the repeated administration of medicinal substances and the protection of better the medicinal substances from degradation, which can increase the capture capacity of the drug and extend its release from the pharmaceutical form.
Collapse
|
37
|
Barberio C, Saez J, Withers A, Nair M, Tamagnini F, Owens RM. Conducting Polymer-ECM Scaffolds for Human Neuronal Cell Differentiation. Adv Healthc Mater 2022; 11:e2200941. [PMID: 35904257 DOI: 10.1002/adhm.202200941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/23/2022] [Indexed: 01/28/2023]
Abstract
3D cell culture formats more closely resemble tissue architecture complexity than 2D systems, which are lacking most of the cell-cell and cell-microenvironment interactions of the in vivo milieu. Scaffold-based systems integrating natural biomaterials are extensively employed in tissue engineering to improve cell survival and outgrowth, by providing the chemical and physical cues of the natural extracellular matrix (ECM). Using the freeze-drying technique, porous 3D composite scaffolds consisting of poly(3,4-ethylene-dioxythiophene) doped with polystyrene sulfonate (PEDOT:PSS), containing ECM components (i.e., collagen, hyaluronic acid, and laminin) are engineered for hosting neuronal cells. The resulting scaffolds exhibit a highly porous microstructure and good conductivity, determined by scanning electron microscopy and electrochemical impedance spectroscopy, respectively. These supports boast excellent mechanical stability and water uptake capacity, making them ideal candidates for cell infiltration. SH-SY5Y human neuroblastoma cells show enhanced cell survival and proliferation in the presence of ECM compared to PEDOT:PSS alone. Whole-cell patch-clamp recordings acquired from differentiated SHSY5Y cells in the scaffolds demonstrate that ECM constituents promote neuronal differentiation in situ. These findings reinforce the usability of 3D conducting supports as engineered highly biomimetic and functional in vitro tissue-like platforms for drug or disease modeling.
Collapse
Affiliation(s)
- Chiara Barberio
- Bioelectronic Systems and Technology group, Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Janire Saez
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, 01006, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, E-48011, Spain
| | - Aimee Withers
- Bioelectronic Systems and Technology group, Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Malavika Nair
- Cambridge Centre for Medical Materials, Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge, CB3 0FS, UK
| | - Francesco Tamagnini
- University of Reading, School of Pharmacy, Hopkins Building, Reading, RG6 6LA, UK
| | - Roisin M Owens
- Bioelectronic Systems and Technology group, Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| |
Collapse
|
38
|
Heng TT, Tey JY, Soon KS, Woo KK. Utilizing Fish Skin of Ikan Belida (Notopterus lopis) as a Source of Collagen: Production and Rheology Properties. Mar Drugs 2022; 20:md20080525. [PMID: 36005530 PMCID: PMC9410226 DOI: 10.3390/md20080525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Collagen hydrogels have been extensively applied in biomedical applications. However, their mechanical properties are insufficient for such applications. Our previous study showed improved mechanical properties when collagen was blended with alginate. The current study aims to analyze the physico-chemical properties of collagen-alginate (CA) films such as swelling, porosity, denaturation temperature (Td), and rheology properties. Collagen was prepared from discarded fish skin of Ikan Belida (Notopterus lopis) that was derived from fish ball manufacturing industries and cross-linked with alginate from brown seaweed (Sargasum polycystum) of a local species as a means to benefit the downstream production of marine industries. CA hydrogels were fabricated with ratios (v/v) of 1:1, 1:4, 3:7, 4:1, and 7:3 respectively. FTIR spectrums of CA film showed an Amide I shift of 1636.12 cm−1 to 1634.64 cm−1, indicating collagen-alginate interactions. SEM images of CA films show a porous structure that varied from pure collagen. DSC analysis shows Td was improved from 61.26 °C (collagen) to 83.11 °C (CA 3:7). CA 4:1 swelled nearly 800% after 48 h, correlated with the of hydrogels porosity. Most CA demonstrated visco-elastic solid characteristics with greater storage modulus (G′) than lost modulus (G″). Shear thinning and non-Newtonian behavior was observed in CA with 0.4% to 1.0% (w/v) CaCl2. CA hydrogels that were derived from discarded materials shows promising potential to serve as a wound dressing or ink for bio printing in the future.
Collapse
Affiliation(s)
- Tzen T. Heng
- Department of Chemical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, Cheras, Kajang 43000, Selangor, Malaysia
| | - Jing Y. Tey
- Department of Mechanical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, Cheras, Kajang 43000, Selangor, Malaysia
| | - Kean S. Soon
- Department of Chemical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, Cheras, Kajang 43000, Selangor, Malaysia
| | - Kwan K. Woo
- Department of Chemical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, Cheras, Kajang 43000, Selangor, Malaysia
- Correspondence:
| |
Collapse
|
39
|
Meganathan I, Sundarapandian A, Shanmugam G, Ayyadurai N. Three-dimensional tailor-made collagen-like proteins hydrogel for tissue engineering applications. BIOMATERIALS ADVANCES 2022; 139:212997. [PMID: 35882145 DOI: 10.1016/j.bioadv.2022.212997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/23/2022] [Accepted: 06/19/2022] [Indexed: 06/15/2023]
Abstract
Despite the potential tunable properties of blank slate collagen-like proteins (CLP), an alternative to animal-originated collagen, assembling them into a stable 3D hydrogel to mimic extracellular matrix is a challenge. To address this constraint, the CLP (without hydroxyproline, CLPpro) and its variants encoding functional unnatural amino acids such as hydroxyproline (CLPhyp) and 3,4-dihydroxyphenylalanine (CLPdopa) were generated through genetic code engineering for 3D hydrogel development. The CLPhyp and CLPdopa were chosen to enhance the intermolecular hydrogen bond interaction through additional hydroxyl moiety and thereby facilitate the self-assembly into a fibrillar network of the hydrogel. Hydrogelation was induced through genipin as a cross-linker, enabling intermolecular cross-linking to form a hydrogel. Spectroscopic and rheological analyses confirmed that CLPpro and its variants maintained native triple-helical structure, which is necessary for its function, and viscoelastic nature of the hydrogels, respectively. Unlike CLPpro, the varients (CLPhyp and CLPdopa) increased pore size formation in the hydrogel scaffold, facilitating 3T3 fibroblast cell interactions. DSC analysis indicated that the stability of the hydrogels got increased upon the genetic incorporation of hydroxyproline (CLPhyp) and dopa (CLPdopa) in CLPpro. In addition, CLPdopa hydrogel was found to be relatively stable against collagenase enzyme compared to CLPpro and CLPhyp. It is the first report on 3D biocompatible hydrogel preparation by tailoring CLP sequence with non-natural amino acids. These next-generation tunable CLP hydrogels open a new venue to design synthetic protein-based biocompatible 3D biomaterials for tissue engineering applications.
Collapse
Affiliation(s)
- Ilamaran Meganathan
- Division of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) - Central Leather Research Institute, Chennai, Tamilnadu, India
| | - Ashokraj Sundarapandian
- Division of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) - Central Leather Research Institute, Chennai, Tamilnadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Ganesh Shanmugam
- Division of Organic and Bioorganic Chemistry, Council of Scientific and Industrial Research (CSIR) - Central Leather Research Institute, Chennai, Tamilnadu, India.
| | - Niraikulam Ayyadurai
- Division of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) - Central Leather Research Institute, Chennai, Tamilnadu, India.
| |
Collapse
|
40
|
Optogenetically Engineered Neurons Differentiated from Human SH-SY5Y Cells Survived and Expressed ChR2 in 3D Hydrogel. Biomedicines 2022; 10:biomedicines10071534. [PMID: 35884839 PMCID: PMC9313127 DOI: 10.3390/biomedicines10071534] [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: 05/17/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 11/25/2022] Open
Abstract
The cases of brain degenerative disease will rise as the human population ages. Current treatments have a transient effect and lack an investigative system that is physiologically relevant for testing. There is evidence suggesting optogenetic stimulation is a potential strategy; however, an in vitro disease and optogenetic model requires a three-dimensional microenvironment. Alginate is a promising material for tissue and optogenetic engineering. Although it is bioinert, alginate hydrogel is transparent and therefore allows optical penetration for stimulation. In this study, alginate was functionalized with arginine-glycine-aspartate acid (RGD) to serve as a 3D platform for encapsulation of human SH-SY5Y cells, which were optogenetically modified and characterized. The RGD-alginate hydrogels were tested for swelling and degradation. Prior to encapsulation, the cells were assessed for neuronal expression and optical-stimulation response. The results showed that RGD-alginate possessed a consistent swelling ratio of 18% on day 7, and degradation remained between 3.7−5% throughout 14 days. Optogenetically modified SH-SY5Y cells were highly viable (>85%) after lentiviral transduction and neuronal differentiation. The cells demonstrated properties of functional neurons, developing beta III tubulin (TuJ1)-positive long neurites, forming neural networks, and expressing vGlut2. Action potentials were produced upon optical stimulation. The neurons derived from human SH-SY5Y cells were successfully genetically modified and encapsulated; they survived and expressed ChR2 in an RGD-alginate hydrogel system.
Collapse
|
41
|
Importance of Matrix Cues on Intervertebral Disc Development, Degeneration, and Regeneration. Int J Mol Sci 2022; 23:ijms23136915. [PMID: 35805921 PMCID: PMC9266338 DOI: 10.3390/ijms23136915] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 01/25/2023] Open
Abstract
Back pain is one of the leading causes of disability worldwide and is frequently caused by degeneration of the intervertebral discs. The discs’ development, homeostasis, and degeneration are driven by a complex series of biochemical and physical extracellular matrix cues produced by and transmitted to native cells. Thus, understanding the roles of different cues is essential for designing effective cellular and regenerative therapies. Omics technologies have helped identify many new matrix cues; however, comparatively few matrix molecules have thus far been incorporated into tissue engineered models. These include collagen type I and type II, laminins, glycosaminoglycans, and their biomimetic analogues. Modern biofabrication techniques, such as 3D bioprinting, are also enabling the spatial patterning of matrix molecules and growth factors to direct regional effects. These techniques should now be applied to biochemically, physically, and structurally relevant disc models incorporating disc and stem cells to investigate the drivers of healthy cell phenotype and differentiation. Such research will inform the development of efficacious regenerative therapies and improved clinical outcomes.
Collapse
|
42
|
Hwang J, An EK, Zhang W, Kim HJ, Eom Y, Jin JO. Dual-functional alginate and collagen–based injectable hydrogel for the treatment of cancer and its metastasis. J Nanobiotechnology 2022; 20:245. [PMID: 35643505 PMCID: PMC9148466 DOI: 10.1186/s12951-022-01458-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/08/2022] [Indexed: 12/19/2022] Open
Abstract
Background Immunotherapies have been gaining attention for the prevention of cancer recurrence and metastasis. Cancer immunotherapy can induce memory cells to target cancer-specific antigens and, thus, selectively kill cancer cells. However, there are difficulties in inducing cancer antigen–specific immunity due to limited knowledge regarding cancer antigens. In this study, we synthesized a dual-functional hydrogel to induce antigen generation and immune activation. Results To elicit a cancer self-antigen–specific immune response, we synthesized an alginate-collagen–based injectable hydrogel, called thermally responsive hydrogel (pTRG), which was incorporated with indocyanine green and the immune stimulator polyinosinic:polycytidylic acid (poly I:C). pTRG was evaluated for its anticancer and anti-metastatic effects against CT-26 carcinoma and 4T1 breast tumor in mice by combining photothermal therapy (PTT) and immunotherapy. Near-infrared (NIR) irradiation promoted temperature elevation in pTRG, consequently exerting a therapeutic effect on mouse tumors. Lung metastasis was prevented in cured CT-26 tumor-injected mice following pTRG treatment via cancer antigen–specific T cell immunity. Moreover, pTRG successfully eliminated the original tumor in 4T1 tumor-bearing mice via PTT and protected them from lung metastasis. To further evaluate the carrier function of TRGs, different types of immunotherapeutic molecules were incorporated into TRGs, which led to the effective elimination of the first CT-26 tumor and the prevention of lung metastasis. Conclusions Our data demonstrate that TRG is a efficient material not only for treating primary tumors but also for preventing metastasis and recurrence.
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01458-x.
Collapse
|
43
|
Xu F, Dawson C, Lamb M, Mueller E, Stefanek E, Akbari M, Hoare T. Hydrogels for Tissue Engineering: Addressing Key Design Needs Toward Clinical Translation. Front Bioeng Biotechnol 2022; 10:849831. [PMID: 35600900 PMCID: PMC9119391 DOI: 10.3389/fbioe.2022.849831] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
Graphical Abstract
Collapse
Affiliation(s)
- Fei Xu
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Chloe Dawson
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Makenzie Lamb
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Eva Mueller
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Evan Stefanek
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC, Canada
| | - Mohsen Akbari
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC, Canada
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
- *Correspondence: Mohsen Akbari, ; Todd Hoare,
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
- *Correspondence: Mohsen Akbari, ; Todd Hoare,
| |
Collapse
|
44
|
Moulahoum H, Ghorbanizamani F, Guler Celik E, Timur S. Nano-Scaled Materials and Polymer Integration in Biosensing Tools. BIOSENSORS 2022; 12:301. [PMID: 35624602 PMCID: PMC9139048 DOI: 10.3390/bios12050301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 12/27/2022]
Abstract
The evolution of biosensors and diagnostic devices has been thriving in its ability to provide reliable tools with simplified operation steps. These evolutions have paved the way for further advances in sensing materials, strategies, and device structures. Polymeric composite materials can be formed into nanostructures and networks of different types, including hydrogels, vesicles, dendrimers, molecularly imprinted polymers (MIP), etc. Due to their biocompatibility, flexibility, and low prices, they are promising tools for future lab-on-chip devices as both manufacturing materials and immobilization surfaces. Polymers can also allow the construction of scaffold materials and 3D structures that further elevate the sensing capabilities of traditional 2D biosensors. This review discusses the latest developments in nano-scaled materials and synthesis techniques for polymer structures and their integration into sensing applications by highlighting their various structural advantages in producing highly sensitive tools that rival bench-top instruments. The developments in material design open a new door for decentralized medicine and public protection that allows effective onsite and point-of-care diagnostics.
Collapse
Affiliation(s)
- Hichem Moulahoum
- Biochemistry Department, Faculty of Science, Ege University, Bornova, 35100 Izmir, Turkey; (H.M.); (F.G.)
| | - Faezeh Ghorbanizamani
- Biochemistry Department, Faculty of Science, Ege University, Bornova, 35100 Izmir, Turkey; (H.M.); (F.G.)
| | - Emine Guler Celik
- Bioengineering Department, Faculty of Science, Ege University, Bornova, 35100 Izmir, Turkey;
| | - Suna Timur
- Biochemistry Department, Faculty of Science, Ege University, Bornova, 35100 Izmir, Turkey; (H.M.); (F.G.)
- Central Research Testing and Analysis Laboratory Research and Application Center, Ege University, Bornova, 35100 Izmir, Turkey
| |
Collapse
|
45
|
Ho TC, Chang CC, Chan HP, Chung TW, Shu CW, Chuang KP, Duh TH, Yang MH, Tyan YC. Hydrogels: Properties and Applications in Biomedicine. Molecules 2022; 27:2902. [PMID: 35566251 PMCID: PMC9104731 DOI: 10.3390/molecules27092902] [Citation(s) in RCA: 204] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 12/19/2022] Open
Abstract
Hydrogels are crosslinked polymer chains with three-dimensional (3D) network structures, which can absorb relatively large amounts of fluid. Because of the high water content, soft structure, and porosity of hydrogels, they closely resemble living tissues. Research in recent years shows that hydrogels have been applied in various fields, such as agriculture, biomaterials, the food industry, drug delivery, tissue engineering, and regenerative medicine. Along with the underlying technology improvements of hydrogel development, hydrogels can be expected to be applied in more fields. Although not all hydrogels have good biodegradability and biocompatibility, such as synthetic hydrogels (polyvinyl alcohol, polyacrylamide, polyethylene glycol hydrogels, etc.), their biodegradability and biocompatibility can be adjusted by modification of their functional group or incorporation of natural polymers. Hence, scientists are still interested in the biomedical applications of hydrogels due to their creative adjustability for different uses. In this review, we first introduce the basic information of hydrogels, such as structure, classification, and synthesis. Then, we further describe the recent applications of hydrogels in 3D cell cultures, drug delivery, wound dressing, and tissue engineering.
Collapse
Affiliation(s)
- Tzu-Chuan Ho
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-C.H.); (C.-W.S.)
| | - Chin-Chuan Chang
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Electrical Engineering, I-Shou University, Kaohsiung 840, Taiwan
| | - Hung-Pin Chan
- Department of Nuclear Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan;
| | - Tze-Wen Chung
- Biomedical Engineering Research and Development Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Chih-Wen Shu
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-C.H.); (C.-W.S.)
| | - Kuo-Pin Chuang
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan;
| | - Tsai-Hui Duh
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Hui Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Center of General Education, Shu-Zen Junior College of Medicine and Management, Kaohsiung 821, Taiwan
| | - Yu-Chang Tyan
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-C.H.); (C.-W.S.)
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan;
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
46
|
Kim S, Lee H, Kim JA, Park TH. Prevention of collagen hydrogel contraction using polydopamine-coating and alginate outer shell increases cell contractile force. BIOMATERIALS ADVANCES 2022; 136:212780. [PMID: 35929298 DOI: 10.1016/j.bioadv.2022.212780] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/07/2022] [Accepted: 03/25/2022] [Indexed: 06/15/2023]
Abstract
Collagen is the most abundant protein in the extracellular matrix of mammals and has a great effect on various cell behaviors including adhesion, differentiation, and migration. However, it is difficult to utilize collagen gel as a physical scaffold in vitro because of its severe contraction. Decrease in the overall hydrogel volume induces changes in cell distribution, and mass transfer within the gel. Uncontrolled mechanical and physiological factors in the fibrous matrix result in uncontrolled cell behaviors in the surrounding cells. In this study, two strategies were used to minimize the contraction of collagen gel. A disk-shaped frame made of polydopamine-coated polydimethylsiloxane (PDMS) prevented horizontal contraction at the edge of the hydrogel. The sequentially cross-linked collagen gel with alginate outer shell (CA-shell) structure inhibited the vertical gel contraction. The combined method synergistically prevented the hydrogel from shrinkage in long-term 3D cell culture. We observed the shift in balance of differentiation from adipogenesis to osteogenesis in mesenchymal stem cells under the environment where gel contraction was prevented, and confirmed that this phenomenon is closely associated with the mechanotransduction based on Yes-associated protein (YAP) localization. Development of this contraction inhibition platform made it possible to investigate the influence of regulation of cellular microenvironments. The physical properties of the hydrogel fabricated in this study were similar to that of pure collagen gel but completely changed the cell behavior within the gel by inhibition of gel contraction. The platform can be used to broaden our understanding of the fundamental mechanism underlying cell-matrix interactions and reproduce extracellular matrix in vivo.
Collapse
Affiliation(s)
- Seulha Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Haein Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju, Chungbuk 28119, Republic of Korea.
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea; Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea; BioMAX/N-Bio Institute, Institute of BioEngineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
47
|
Yan M, An X, Jiang Z, Duan S, Wang A, Zhao X, Li Y. Effects of cross-linking with EDC/NHS and genipin on characterizations of self-assembled fibrillar gel prepared from tilapia collagen and alginate. Polym Degrad Stab 2022. [DOI: 10.1016/j.polymdegradstab.2022.109929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
48
|
Antunes M, Bonani W, Reis RL, Migliaresi C, Ferreira H, Motta A, Neves NM. Development of alginate-based hydrogels for blood vessel engineering. BIOMATERIALS ADVANCES 2022; 134:112588. [PMID: 35525739 DOI: 10.1016/j.msec.2021.112588] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/09/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022]
Abstract
Vascular diseases are among the primary causes of death worldwide. In serious conditions, replacement of the damaged vessel is required. Autologous grafts are preferred, but their limited availability and difficulty of the harvesting procedures favour synthetic alternatives' use. However, as synthetic grafts may present significant drawbacks, tissue engineering-based solutions are proposed. Herein, tubular hydrogels of alginate combined with collagen type I and/or silk fibroin were prepared by ionotropic gelation using gelatin hydrogel sacrificial moulds loaded with calcium ions (Ca2+). The time of exposure of alginate solutions to Ca2+-loaded gelatin was used to control the wall thickness of the hydrogels (0.47 ± 0.10 mm-1.41 ± 0.21 mm). A second crosslinking step with barium chloride prevented their degradation for a 14 day period and improved mechanical properties by two-fold. Protein leaching tests showed that collagen type I, unlike silk fibroin, was strongly incorporated in the hydrogels. The presence of silk fibroin in the alginate matrix, containing or not collagen, did not significantly improve hydrogels' properties. Conversely, hydrogels enriched only with collagen were able to better support EA.hy926 and MRC-5 cells' growth and characteristic phenotype. These results suggest that a two-step crosslinking procedure combined with the use of collagen type I allow for producing freestanding vascular substitutes with tuneable properties in terms of size, shape and wall thickness.
Collapse
Affiliation(s)
- Margarida Antunes
- 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, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Walter Bonani
- Department of Industrial Engineering, University of Trento, via Sommarive, 9, 38123 Trento, Italy; BIOtech Research Centre, University of Trento, via delle Regole 101, 38123 Mattarello, Trento, Italy
| | - 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, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Claudio Migliaresi
- Department of Industrial Engineering, University of Trento, via Sommarive, 9, 38123 Trento, Italy; BIOtech Research Centre, University of Trento, via delle Regole 101, 38123 Mattarello, Trento, Italy
| | - Helena Ferreira
- 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, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Antonella Motta
- Department of Industrial Engineering, University of Trento, via Sommarive, 9, 38123 Trento, Italy; BIOtech Research Centre, University of Trento, via delle Regole 101, 38123 Mattarello, Trento, Italy
| | - Nuno M Neves
- 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, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
49
|
Bioengineered models of Parkinson's disease using patient-derived dopaminergic neurons exhibit distinct biological profiles in a 3D microenvironment. Cell Mol Life Sci 2022; 79:78. [PMID: 35044538 PMCID: PMC8908880 DOI: 10.1007/s00018-021-04047-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/05/2021] [Accepted: 11/17/2021] [Indexed: 01/21/2023]
Abstract
Three-dimensional (3D) in vitro culture systems using human induced pluripotent stem cells (hiPSCs) are useful tools to model neurodegenerative disease biology in physiologically relevant microenvironments. Though many successful biomaterials-based 3D model systems have been established for other neurogenerative diseases, such as Alzheimer's disease, relatively few exist for Parkinson's disease (PD) research. We employed tissue engineering approaches to construct a 3D silk scaffold-based platform for the culture of hiPSC-dopaminergic (DA) neurons derived from healthy individuals and PD patients harboring LRRK2 G2019S or GBA N370S mutations. We then compared results from protein, gene expression, and metabolic analyses obtained from two-dimensional (2D) and 3D culture systems. The 3D platform enabled the formation of dense dopamine neuronal network architectures and developed biological profiles both similar and distinct from 2D culture systems in healthy and PD disease lines. PD cultures developed in 3D platforms showed elevated levels of α-synuclein and alterations in purine metabolite profiles. Furthermore, computational network analysis of transcriptomic networks nominated several novel molecular interactions occurring in neurons from patients with mutations in LRRK2 and GBA. We conclude that the brain-like 3D system presented here is a realistic platform to interrogate molecular mechanisms underlying PD biology.
Collapse
|
50
|
Ozgun A, Lomboni D, Arnott H, Staines WA, Woulfe J, Variola F. Biomaterial-based strategies for in vitro neural models. Biomater Sci 2022; 10:1134-1165. [PMID: 35023513 DOI: 10.1039/d1bm01361k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In vitro models have been used as a complementary tool to animal studies in understanding the nervous system's physiological mechanisms and pathological disorders, while also serving as platforms to evaluate the safety and efficiency of therapeutic candidates. Following recent advances in materials science, micro- and nanofabrication techniques and cell culture systems, in vitro technologies have been rapidly gaining the potential to bridge the gap between animal and clinical studies by providing more sophisticated models that recapitulate key aspects of the structure, biochemistry, biomechanics, and functions of human tissues. This was made possible, in large part, by the development of biomaterials that provide cells with physicochemical features that closely mimic the cellular microenvironment of native tissues. Due to the well-known material-driven cellular response and the importance of mimicking the environment of the target tissue, the selection of optimal biomaterials represents an important early step in the design of biomimetic systems to investigate brain structures and functions. This review provides a comprehensive compendium of commonly used biomaterials as well as the different fabrication techniques employed for the design of neural tissue models. Furthermore, the authors discuss the main parameters that need to be considered to develop functional platforms not only for the study of brain physiological functions and pathological processes but also for drug discovery/development and the optimization of biomaterials for neural tissue engineering.
Collapse
Affiliation(s)
- Alp Ozgun
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - David Lomboni
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - Hallie Arnott
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - William A Staines
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - John Woulfe
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada.,The Ottawa Hospital, Ottawa, Canada
| | - Fabio Variola
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada.,The Ottawa Hospital, Ottawa, Canada.,Children's Hospital of Eastern Ontario (CHEO), Ottawa, Canada
| |
Collapse
|