1
|
Zhou Z, Zhang Y, Zeng Y, Yang D, Mo J, Zheng Z, Zhang Y, Xiao P, Zhong X, Yan W. Effects of Nanomaterials on Synthesis and Degradation of the Extracellular Matrix. ACS NANO 2024; 18:7688-7710. [PMID: 38436232 DOI: 10.1021/acsnano.3c09954] [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: 03/05/2024]
Abstract
Extracellular matrix (ECM) remodeling is accompanied by the continuous synthesis and degradation of the ECM components. This dynamic process plays an important role in guiding cell adhesion, migration, proliferation, and differentiation, as well as in tissue development, body repair, and maintenance of homeostasis. Nanomaterials, due to their photoelectric and catalytic properties and special structure, have garnered much attention in biomedical fields for use in processes such as tissue engineering and disease treatment. Nanomaterials can reshape the cell microenvironment by changing the synthesis and degradation of ECM-related proteins, thereby indirectly changing the behavior of the surrounding cells. This review focuses on the regulatory role of nanomaterials in the process of cell synthesis of different ECM-related proteins and extracellular protease. We discuss influencing factors and possible related mechanisms of nanomaterials in ECM remodeling, which may provide different insights into the design and development of nanomaterials for the treatment of ECM disorder-related diseases.
Collapse
Affiliation(s)
- Zhiyan Zhou
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510260, China
| | - Yuting Zeng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dehong Yang
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiayao Mo
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ziting Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuxin Zhang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ping Xiao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xincen Zhong
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenjuan Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
2
|
Chen YM, Tokoda C, Tabata Y. Cell culture design for homogeneous proliferation of cells in three-dimensional nonwoven polymer scaffolds. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:1-15. [PMID: 37773043 DOI: 10.1080/09205063.2023.2265623] [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: 08/01/2023] [Accepted: 09/21/2023] [Indexed: 09/30/2023]
Abstract
The objective of this study is to establish strategies to uniformly proliferate cells in a three-dimensional nonwoven polyethylene terephthalate (PET)/ethylene vinyl alcohol (EVOH) scaffold by simple adjustments in seeding and culture methods and the scaffold design. The combined dynamic and static seeding (intermittent agitations at 300 rpm with 1 h interval) resulted in the highest seeding efficiency (71%) comparing to the static and continuous agitating seeding methods. Cell-attached scaffolds were cultivated under different conditions. The stirring culture permitted cells to proliferate to a significantly greater extent than the static or agitating cultures, although faster cell proliferation in the outer region of the scaffold was observed. Next, based on this observation, scaffolds were opened with holes to alleviate the cell aggregation. The effect of hole size and number of scaffolds on the distribution of cells proliferated in the scaffold was evaluated. Two of 1-mm holes showed to be an optimal adjustment to allow cells to proliferate in a homogeneous manner. After 14 days culture, both of the holes were filled by cells proliferated with a fourfold increase in the cell number. The cell viability in the scaffolds was also high upon evaluating the live/dead and 3[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) staining examinations. Different cell types of 3T3-L1, C3H/10T1/2, and KUM6 cells showed similar behavior of cell proliferation and distribution in the scaffold, indicating the applicability of the established procedure. It is concluded that the nonwoven PET/EVOH scaffold serves as a potential cell culture substrate for an efficient cell proliferation.
Collapse
Affiliation(s)
- Yu-Min Chen
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Chihoko Tokoda
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
3
|
Loffet EA, Durel JF, Gao J, Kam R, Lim H, Nerurkar NL. Elastic fibers define embryonic tissue stiffness to enable buckling morphogenesis of the small intestine. Biomaterials 2023; 303:122405. [PMID: 38000151 PMCID: PMC10842730 DOI: 10.1016/j.biomaterials.2023.122405] [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: 07/24/2023] [Revised: 10/22/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023]
Abstract
During embryonic development, tissues must possess precise material properties to ensure that cell-generated forces give rise to the stereotyped morphologies of developing organs. However, the question of how material properties are established and regulated during development remains understudied. Here, we aim to address these broader questions through the study of intestinal looping, a process by which the initially straight intestinal tube buckles into loops, permitting ordered packing within the body cavity. Looping results from elongation of the tube against the constraint of an attached tissue, the dorsal mesentery, which is elastically stretched by the elongating tube to nearly triple its length. This elastic energy storage allows the mesentery to provide stable compressive forces that ultimately buckle the tube into loops. Beginning with a transcriptomic analysis of the mesentery, we identified widespread upregulation of extracellular matrix related genes during looping, including genes related to elastic fiber deposition. Combining molecular and mechanical analyses, we conclude that elastin confers tensile stiffness to the mesentery, enabling its mechanical role in organizing the developing small intestine. These results shed light on the role of elastin as a driver of morphogenesis that extends beyond its more established role in resisting cyclic deformation in adult tissues.
Collapse
Affiliation(s)
- Elise A Loffet
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - John F Durel
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Jenny Gao
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Richard Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Hyunjee Lim
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Nandan L Nerurkar
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
4
|
Wöltje M, Künzelmann L, Belgücan B, Croft AS, Voumard B, Bracher S, Zysset P, Gantenbein B, Cherif C, Aibibu D. Textile Design of an Intervertebral Disc Replacement Device from Silk Yarn. Biomimetics (Basel) 2023; 8:biomimetics8020152. [PMID: 37092404 PMCID: PMC10123607 DOI: 10.3390/biomimetics8020152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 04/25/2023] Open
Abstract
Low back pain is often due to degeneration of the intervertebral discs (IVD). It is one of the most common age- and work-related problems in today's society. Current treatments are not able to efficiently restore the full function of the IVD. Therefore, the aim of the present work was to reconstruct the two parts of the intervertebral disc-the annulus fibrosus (AF) and the nucleus pulposus (NP)-in such a way that the natural structural features were mimicked by a textile design. Silk was selected as the biomaterial for realization of a textile IVD because of its cytocompatibility, biodegradability, high strength, stiffness, and toughness, both in tension and compression. Therefore, an embroidered structure made of silk yarn was developed that reproduces the alternating fiber structure of +30° and -30° fiber orientation found in the AF and mimics its lamellar structure. The developed embroidered ribbons showed a tensile strength that corresponded to that of the natural AF. Fiber additive manufacturing with 1 mm silk staple fibers was used to replicate the fiber network of the NP and generate an open porous textile 3D structure that may serve as a reinforcement structure for the gel-like NP.
Collapse
Affiliation(s)
- Michael Wöltje
- Institute of Textile Machinery and High-Performance Material Technology, Technische Universität Dresden, 01602 Dresden, Germany
| | - Liesa Künzelmann
- Institute of Textile Machinery and High-Performance Material Technology, Technische Universität Dresden, 01602 Dresden, Germany
| | - Basak Belgücan
- Institute of Textile Machinery and High-Performance Material Technology, Technische Universität Dresden, 01602 Dresden, Germany
| | - Andreas S Croft
- Tissue Engineering for Orthopaedic and Mechanobiology, Bone and Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, 3008 Bern, Switzerland
| | - Benjamin Voumard
- ARTORG Center for Biomedical Engineering Research, University of Bern, 3008 Bern, Switzerland
| | - Stefan Bracher
- ARTORG Center for Biomedical Engineering Research, University of Bern, 3008 Bern, Switzerland
| | - Philippe Zysset
- ARTORG Center for Biomedical Engineering Research, University of Bern, 3008 Bern, Switzerland
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedic and Mechanobiology, Bone and Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, 3008 Bern, Switzerland
- Department of Orthopedic Surgery and Traumatology, Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Chokri Cherif
- Institute of Textile Machinery and High-Performance Material Technology, Technische Universität Dresden, 01602 Dresden, Germany
| | - Dilbar Aibibu
- Institute of Textile Machinery and High-Performance Material Technology, Technische Universität Dresden, 01602 Dresden, Germany
| |
Collapse
|
5
|
Sun J, Chan YT, Ho KWK, Zhang L, Bian L, Tuan RS, Jiang Y. "Slow walk" mimetic tensile loading maintains human meniscus tissue resident progenitor cells homeostasis in photocrosslinked gelatin hydrogel. Bioact Mater 2023; 25:256-272. [PMID: 36825224 PMCID: PMC9941420 DOI: 10.1016/j.bioactmat.2023.01.025] [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: 11/01/2022] [Revised: 01/14/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Meniscus, the cushion in knee joint, is a load-bearing tissue that transfers mechanical forces to extracellular matrix (ECM) and tissue resident cells. The mechanoresponse of human tissue resident stem/progenitor cells in meniscus (hMeSPCs) is significant to tissue homeostasis and regeneration but is not well understood. This study reports that a mild cyclic tensile loading regimen of ∼1800 loads/day on hMeSPCs seeded in 3-dimensional (3D) photocrosslinked gelatin methacryloyl (GelMA) hydrogel is critical in maintaining cellular homeostasis. Experimentally, a "slow walk" biomimetic cyclic loading regimen (10% tensile strain, 0.5 Hz, 1 h/day, up to 15 days) is applied to hMeSPCs encapsulated in GelMA hydrogel with a magnetic force-controlled loading actuator. The loading significantly increases cell differentiation and fibrocartilage-like ECM deposition without affecting cell viability. Transcriptomic analysis reveals 332 mechanoresponsive genes, clustered into cell senescence, mechanical sensitivity, and ECM dynamics, associated with interleukins, integrins, and collagens/matrix metalloproteinase pathways. The cell-GelMA constructs show active ECM remodeling, traced using a green fluorescence tagged (GFT)-GelMA hydrogel. Loading enhances nascent pericellular matrix production by the encapsulated hMeSPCs, which gradually compensates for the hydrogel loss in the cultures. These findings demonstrate the strong tissue-forming ability of hMeSPCs, and the importance of mechanical factors in maintaining meniscus homeostasis.
Collapse
Key Words
- 3D cell-based constructs
- 3D, Three-dimensional
- BMSCs, Bone marrow derived mesenchymal stem cells
- Biomimetic cyclic loading
- CFUs, Colony forming units
- Col I, Collagen type I
- Col II, Collagen type II
- DS, Degree of substitution
- ECM, Extracellular matrix
- Extracellular matrix
- GAGs, Glycosaminoglycans
- GFT-GelMA, Green fluorescence-tagged GelMA
- GelMA hydrogel
- GelMA, Gelatin methacryloyl
- Human meniscus progenitor cells
- MeHA, Methacrylated hyaluronic acid
- PCM, Pericellular matrix
- PI, Propidium iodide
- PPI, Protein-protein interaction
- hMeSPCs, Human meniscus stem/progenitor cells
Collapse
Affiliation(s)
- Jing Sun
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Yau Tsz Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Ki Wai Kevin Ho
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, And Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
| | - Li Zhang
- Department of Mechanical and Automation Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Liming Bian
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China,Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China,Corresponding author. Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China.
| | - Yangzi Jiang
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China,Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China,Corresponding author. Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China.
| |
Collapse
|
6
|
Nanotechnology in tissue engineering and regenerative medicine. KOREAN J CHEM ENG 2023. [DOI: 10.1007/s11814-022-1363-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
7
|
Sun L, Li X, Yang T, Lu T, Du P, Jing C, Chen Z, Lin F, Zhao G, Zhao L. Construction of spider silk protein small-caliber tissue engineering vascular grafts based on dynamic culture and its performance evaluation. J Biomed Mater Res A 2023; 111:71-87. [PMID: 36129207 DOI: 10.1002/jbm.a.37447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/03/2022] [Accepted: 09/07/2022] [Indexed: 11/12/2022]
Abstract
Tissue engineering is an alternative method for preparing small-caliber (<6 mm) vascular grafts. Dynamic mechanical conditioning is being researched as a method to improve mechanical properties of tissue engineered blood vessels. This method attempts to induce unique reaction in implanted cells that regenerate the matrix around them, thereby improving the overall mechanical stability of the grafts. In this study, we used a bioreactor to seed endothelial cells and smooth muscle cells into the inner and outer layers of the electrospun spider silk protein scaffold respectively to construct vascular grafts. The cell proliferation, mechanical properties, blood compatibility and other indicators of the vascular grafts were characterized in vitro. Furthermore, the vascular grafts were implanted in Sprague Dawley rats, and the vascular grafts' patency, extracellular matrix formation, and inflammatory response were evaluated in vivo. We aimed to construct spider silk protein vascular grafts with the potential for in vivo implantation by using a pulsating flow bioreactor. The results showed that, when compared with the static culture condition, the dynamic culture condition improved cell proliferation on vascular scaffolds and enhanced mechanical function of vascular scaffolds. In vivo experiments also showed that the dynamic culture of vascular grafts was more beneficial for the extracellular matrix deposition and anti-thrombogenesis, as well as reducing the inflammatory response of vascular grafts. In conclusion, dynamic mechanical conditioning aid in the resolution of challenges impeding the application of electrospun scaffolds and have the potential to construct small-caliber blood vessels with regenerative function for cardiovascular tissue repair.
Collapse
Affiliation(s)
- Lulu Sun
- College of Life Science and Technology, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Xiafei Li
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Tuo Yang
- College of Life Science and Technology, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China.,Department of Cardiothoracic Surgery, Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Tian Lu
- College of Life Science and Technology, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China.,Department of Cardiothoracic Surgery, Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Pengchong Du
- College of Life Science and Technology, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China.,Department of Cardiothoracic Surgery, Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Changqin Jing
- College of Life Science and Technology, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Zhigang Chen
- Henan Engineering Research Center for Mitochondrion Biomedical of Heart, Henan Joint International Research Laboratory of Cardiovascular Injury and Repair, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Fei Lin
- Henan Engineering Research Center for Mitochondrion Biomedical of Heart, Henan Joint International Research Laboratory of Cardiovascular Injury and Repair, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Guoan Zhao
- Henan Engineering Research Center for Mitochondrion Biomedical of Heart, Henan Joint International Research Laboratory of Cardiovascular Injury and Repair, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Liang Zhao
- College of Life Science and Technology, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China.,Henan Engineering Research Center for Mitochondrion Biomedical of Heart, Henan Joint International Research Laboratory of Cardiovascular Injury and Repair, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China.,The Central Lab, The Third People Hospital of Datong, Datong, China
| |
Collapse
|
8
|
Badaraev AD, Sidelev DV, Kozelskaya AI, Bolbasov EN, Tran TH, Nashchekin AV, Malashicheva AB, Rutkowski S, Tverdokhlebov SI. Surface Modification of Electrospun Bioresorbable and Biostable Scaffolds by Pulsed DC Magnetron Sputtering of Titanium for Gingival Tissue Regeneration. Polymers (Basel) 2022; 14:4922. [PMID: 36433049 PMCID: PMC9698656 DOI: 10.3390/polym14224922] [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: 10/25/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, polymer scaffolds were fabricated from biodegradable poly(lactide-co-glycolide) (PLGA) and from non-biodegradable vinylidene fluoride-tetrafluoroethylene (VDF-TeFE) by electrospinning. These polymer scaffolds were subsequently surface-modified by sputtering titanium targets in an argon atmosphere. Direct current pulsed magnetron sputtering was applied to prevent a significant influence of discharge plasma on the morphology and mechanical properties of the nonwoven polymer scaffolds. The scaffolds with initially hydrophobic properties show higher hydrophilicity and absorbing properties after surface modification with titanium. The surface modification by titanium significantly increases the cell adhesion of both the biodegradable and the non-biodegradable scaffolds. Immunocytochemistry investigations of human gingival fibroblast cells on the surface-modified scaffolds indicate that a PLGA scaffold exhibits higher cell adhesion than a VDF-TeFE scaffold.
Collapse
Affiliation(s)
- Arsalan D. Badaraev
- Weinberg Research Center, School of Nuclear Science & Engineering, National Research Tomsk Polytechnic University, 30, Lenin Avenue, 634050 Tomsk, Russia
| | - Dmitrii V. Sidelev
- Weinberg Research Center, School of Nuclear Science & Engineering, National Research Tomsk Polytechnic University, 30, Lenin Avenue, 634050 Tomsk, Russia
| | - Anna I. Kozelskaya
- Weinberg Research Center, School of Nuclear Science & Engineering, National Research Tomsk Polytechnic University, 30, Lenin Avenue, 634050 Tomsk, Russia
| | - Evgeny N. Bolbasov
- Weinberg Research Center, School of Nuclear Science & Engineering, National Research Tomsk Polytechnic University, 30, Lenin Avenue, 634050 Tomsk, Russia
| | - Tuan-Hoang Tran
- Weinberg Research Center, School of Nuclear Science & Engineering, National Research Tomsk Polytechnic University, 30, Lenin Avenue, 634050 Tomsk, Russia
| | | | | | - Sven Rutkowski
- Weinberg Research Center, School of Nuclear Science & Engineering, National Research Tomsk Polytechnic University, 30, Lenin Avenue, 634050 Tomsk, Russia
| | - Sergei I. Tverdokhlebov
- Weinberg Research Center, School of Nuclear Science & Engineering, National Research Tomsk Polytechnic University, 30, Lenin Avenue, 634050 Tomsk, Russia
| |
Collapse
|
9
|
Li Y, Zha Y, Hu W, Chen J, Liu S, Zhang S, Wang J. Monoporous Microsphere as a Dynamically Movable Drug Carrier for Osteoporotic Bone Remodeling. Adv Healthc Mater 2022:e2201242. [PMID: 35948299 DOI: 10.1002/adhm.202201242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/11/2022] [Indexed: 11/06/2022]
Abstract
To repair systematically osteoporotic bone defects, it is significant to take effort on both the diminishment of osteoporosis and the enhancement of bone regeneration. Herein, a specifically monoporous microsphere carrier encapsulating dosage-sensitive and short half-time parathyroid hormone (PTH) has been constructed to tackle the issue. Compared with conventional microsphere carriers involving compact, porous, and mesoporous microspheres, the monoporous microsphere is desirable to achieve precisely in-situ delivery and to minimize topical accumulation. Our findings show that the PTH loaded inside MPMs can be gradually released from the single hole of MPMs to improve the initial drug concentration. Also, the MPMs can self-shift with the daily movement of experimental animals to effectively reduce the topical aggregation of released drugs in vitro. In vivo evaluation further confirms that the implant of MPMs-PTH plays a dual role in stimulating the regenerative repair of the cranial defect and relieving osteoporosis in the whole body. Consequently, our current work develops a dynamically movable drug delivery system to achieve precisely in-situ delivery, minimize topical accumulation, and systematically repair osteoporotic bone defects. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yawu Li
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Yao Zha
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Weikang Hu
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China.,School of Materials Science and Engineering, Hubei University, Wuhan, Hubei, 430062, P. R. China
| | - Jia Chen
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Shuaibing Liu
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Shengmin Zhang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Jianglin Wang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.,NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| |
Collapse
|
10
|
Chu G, Zhang W, Han F, Li K, Liu C, Wei Q, Wang H, Liu Y, Han F, Li B. The role of microenvironment in stem cell-based regeneration of intervertebral disc. Front Bioeng Biotechnol 2022; 10:968862. [PMID: 36017350 PMCID: PMC9395990 DOI: 10.3389/fbioe.2022.968862] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/18/2022] [Indexed: 01/07/2023] Open
Abstract
Regenerative medicine for intervertebral disc (IVD) disease, by utilizing chondrocytes, IVD cells, and stem cells, has progressed to clinical trials in the treatment of back pain, and has been studied in various animal models of disc degeneration in the past decade. Stem cells exist in their natural microenvironment, which provides vital dynamic physical and chemical signals for their survival, proliferation and function. Long-term survival, function and fate of mesenchymal stem cells (MSCs) depend on the microenvironment in which they are transplanted. However, the transplanted MSCs and the endogenous disc cells were influenced by the complicated microenvironment in the degenerating disc with the changes of biochemical and biophysical components. It is important to understand how the MSCs and endogenous disc cells survive and thrive in the harsh microenvironment of the degenerative disc. Furthermore, materials containing stem cells and their natural microenvironment have good clinical effects. However, the implantation of tissue engineering IVD (TE-IVD) cannot provide a complete and dynamic microenvironment for MSCs. IVD graft substitutes may need further improvement to provide the best engineered MSC microenvironment. Additionally, the IVD progenitor cells inside the stem cell niches have been regarded as popular graft cells for IVD regeneration. However, it is still unclear whether actual IVD progenitor cells exist in degenerative spinal conditions. Therefore, the purpose of this review is fourfold: to discuss the presence of endogenous stem cells; to review and summarize the effects of the microenvironment in biological characteristics of MSC, especially those from IVD; to explore the feasibility and prospects of IVD graft substitutes and to elaborate state of the art in the use of MSC transplantation for IVD degeneration in vivo as well as their clinical application.
Collapse
Affiliation(s)
- Genglei Chu
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Weidong Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Feng Han
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Kexin Li
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Chengyuan Liu
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Qiang Wei
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Huan Wang
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yijie Liu
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Fengxuan Han
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Bin Li
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Suzhou Medical College, Soochow University, Suzhou, China
| |
Collapse
|
11
|
Enthesis Healing Is Dependent on Scaffold Interphase Morphology—Results from a Rodent Patellar Model. Cells 2022; 11:cells11111752. [PMID: 35681447 PMCID: PMC9179925 DOI: 10.3390/cells11111752] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 01/27/2023] Open
Abstract
The use of multiphasic scaffolds to treat injured tendon-to-bone entheses has shown promising results in vitro. Here, we used two versions of a biphasic silk fibroin scaffold to treat an enthesis defect created in a rat patellar model in vivo. One version presented a mixed transition between the bony and the tendon end of the construct (S-MT) while this transition was abrupt in the second version (S-AT). At 12 weeks after surgery, the S-MT scaffold promoted better healing of the injured enthesis, with minimal undesired ossification of the insertion area. The expression of tenogenic and chondrogenic markers was sustained for longer in the S-MT-treated group and the tangent modulus of the S-MT-treated samples was similar to the native tissue at 12 weeks while that of the S-AT-treated enthesis was lower. Our study highlights the important role of the transition zone of multiphasic scaffolds in the treatment of complex interphase tissues such as the tendon-to-bone enthesis.
Collapse
|
12
|
Mao B, Zhang Z, Lai S, Zhang K, Li J, Fu W. Demineralized Cortical Bone Matrix Augmented With Peripheral Blood-Derived Mesenchymal Stem Cells for Rabbit Medial Meniscal Reconstruction. Front Bioeng Biotechnol 2022; 10:855103. [PMID: 35573229 PMCID: PMC9091599 DOI: 10.3389/fbioe.2022.855103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/12/2022] [Indexed: 11/24/2022] Open
Abstract
Tissue engineering is a promising treatment strategy for meniscal regeneration after meniscal injury. However, existing scaffold materials and seed cells still have many disadvantages. The objective of the present study is to explore the feasibility of peripheral blood-derived mesenchymal stem cells (PBMSCs) augmented with demineralized cortical bone matrix (DCBM) pretreated with TGF-β3 as a tissue-engineered meniscus graft and the repair effect. PBMSCs were collected from rabbit peripheral blood and subjected to three-lineage differentiation and flow cytometry identification. DCBM was prepared by decalcification, decellularization, and cross-linking rabbit cortical bone. Various characteristics such as biomechanical properties, histological characteristics, microstructure and DNA content were characterized. The cytotoxicity and the effects of DCBM on the adhesion and migration of PBMSCs were evaluated separately. The meniscus-forming ability of PBMSCs/DCBM complex in vitro induced by TGF-β3 was also evaluated at the molecular and genetic levels, respectively. Eventually, the present study evaluated the repair effect and cartilage protection effect of PBMSCs/DCBM as a meniscal graft in a rabbit model of medial meniscal reconstruction in 3 and 6 months. The results showed PBMSCs positively express CD29 and CD44, negatively express CD34 and CD45, and have three-lineage differentiation ability, thus can be used as tissue engineering meniscus seed cells. After the sample procedure, the cell and DNA contents of DCBM decreased, the tensile modulus did not decrease significantly, and the DCBM had a pore structure and no obvious cytotoxicity. PBMSCs could adhere and grow on the scaffold. Under induction of TGF-β3, PBMSCs/DCBM composites expressed glycosaminoglycan (GAG), and the related gene expression also increased. The results of the in vivo experiments that the PBMSCs/DCBM group had a better repair effect than the DCBM group and the control group at both 12 and 24 weeks, and the protective effect on cartilage was also better. Therefore, the application of DCBM augmented with PBMSCs for meniscus injury treatment is a preferred option for tissue-engineered meniscus.
Collapse
Affiliation(s)
- Beini Mao
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Zhong Zhang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Department of Orthopaedics, No.3 People’s Hospital of Chengdu, Chengdu, China
| | - Sike Lai
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Kaibo Zhang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Weili Fu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Delaine-Smith RM, Hann AJ, Green NH, Reilly GC. Electrospun Fiber Alignment Guides Osteogenesis and Matrix Organization Differentially in Two Different Osteogenic Cell Types. Front Bioeng Biotechnol 2021; 9:672959. [PMID: 34760876 PMCID: PMC8573409 DOI: 10.3389/fbioe.2021.672959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/08/2021] [Indexed: 11/18/2022] Open
Abstract
Biomimetic replication of the structural anisotropy of musculoskeletal tissues is important to restore proper tissue mechanics and function. Physical cues from the local micro-environment, such as matrix fiber orientation, may influence the differentiation and extracellular matrix (ECM) organization of osteogenic progenitor cells. This study investigates how scaffold fiber orientation affects the behavior of mature and progenitor osteogenic cells, the influence on secreted mineralized-collagenous matrix organization, and the resulting construct mechanical properties. Gelatin-coated electrospun poly(caprolactone) fibrous scaffolds were fabricated with either a low or a high degree of anisotropy and cultured with mature osteoblasts (MLO-A5s) or osteogenic mesenchymal progenitor cells (hES-MPs). For MLO-A5 cells, alkaline phosphatase (ALP) activity was highest, and more calcium-containing matrix was deposited onto aligned scaffolds. In contrast, hES-MPs, osteogenic mesenchymal progenitor cells, exhibited higher ALP activity, collagen, and calcium deposition on randomly orientated fibers compared with aligned counterparts. Deposited matrix was isotropic on random fibrous scaffolds, whereas a greater degree of anisotropy was observed in aligned fibrous constructs, as confirmed by second harmonic generation (SHG) and scanning electron microscope (SEM) imaging. This resulted in anisotropic mechanical properties on aligned constructs. This study indicates that mineralized-matrix deposition by osteoblasts can be controlled by scaffold alignment but that the early stages of osteogenesis may not benefit from culture on orientated scaffolds.
Collapse
Affiliation(s)
- Robin M. Delaine-Smith
- Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield, United Kingdom
| | - Alice Jane Hann
- Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Materials Science and Engineering, INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Nicola H. Green
- Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Materials Science and Engineering, INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Gwendolen Clair Reilly
- Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Materials Science and Engineering, INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
14
|
Garcia Garcia A, Perot JB, Beldjilali-Labro M, Dermigny Q, Naudot M, Le Ricousse S, Legallais C, Bedoui F. Monitoring mechanical stimulation for optimal tendon tissue engineering: A mechanical and biological multiscale study. J Biomed Mater Res A 2021; 109:1881-1892. [PMID: 33871170 DOI: 10.1002/jbm.a.37180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022]
Abstract
To understand the effect of mechanical stimulation on cell response, bone marrow stromal cells were cultured on electrospun scaffolds under two distinct mechanical conditions (static and dynamic). Comparison between initial and final mechanical and biological properties of the cell-constructs were conducted over 14 days for both culturing conditions. As a result, mechanically stimulated constructs, in contrast to their static counterparts, showed evident mechanical-induced cell orientation, an effective aligned collagen and tenomodulin extracellular matrix. This orientation provides clues on the importance of mechanical stimulation to induce a tendon-like differentiation. In addition, cell and collagen orientation lead to enhanced storage modulus observed under dynamic stimulation. Altogether mechanical stimulation lead to (a) cell and matrix orientation through the sense of the stretch and (b) a dominant elastic response in the cell-constructs with a minor contribution of the viscosity in the global mechanical behavior. Such a correlation could help in further studies to better understand the effect of mechanical stimulation in tissue engineering.
Collapse
Affiliation(s)
- Alejandro Garcia Garcia
- CNRS, UMR 7338 Laboratory of Biomechanics and Bioengineering, Sorbonne Universités, Université de Technologie de Compiègne, Compiegne, France
| | - Jean-Baptiste Perot
- CNRS, UMR 7338 Laboratory of Biomechanics and Bioengineering, Sorbonne Universités, Université de Technologie de Compiègne, Compiegne, France
| | - Megane Beldjilali-Labro
- CNRS, UMR 7338 Laboratory of Biomechanics and Bioengineering, Sorbonne Universités, Université de Technologie de Compiègne, Compiegne, France
| | - Quentin Dermigny
- CNRS, UMR 7338 Laboratory of Biomechanics and Bioengineering, Sorbonne Universités, Université de Technologie de Compiègne, Compiegne, France
| | - Marie Naudot
- EA4666-LNPC-Immunologie Therapie Cellulaire Hématologie Cancers, CURS, Hopital Sud Avenue René Laennec, Salouel, France
| | - Sophie Le Ricousse
- EA4666-LNPC-Immunologie Therapie Cellulaire Hématologie Cancers, CURS, Hopital Sud Avenue René Laennec, Salouel, France
| | - Cecile Legallais
- CNRS, UMR 7338 Laboratory of Biomechanics and Bioengineering, Sorbonne Universités, Université de Technologie de Compiègne, Compiegne, France
| | - Fahmi Bedoui
- FRE CNRS 2012 Roberval Laboratory for Mechanics, Sorbonne Universités, Université de Technologie de Compiègne, Compiègne, France
| |
Collapse
|
15
|
Page MI, Linde PE, Puttlitz CM. High throughput computational evaluation of how scaffold architecture, material selection, and loading modality influence the cellular micromechanical environment in tissue engineering strategies. JOR Spine 2021; 4:e1152. [PMID: 34611587 PMCID: PMC8479525 DOI: 10.1002/jsp2.1152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND In tissue engineering (TE) strategies, cell processes are regulated by mechanical stimuli. Although TE scaffolds have been developed to replicate tissue-level mechanical properties, it is intractable to experimentally measure and prescribe the cellular micromechanical environment (CME) generated within these constructs. Accordingly, this study aimed to fill this lack of understanding by modeling the CME in TE scaffolds using the finite element method. METHODS A repeating unit of composite fiber scaffold for annulus fibrosus (AF) repair with a fibrin hydrogel matrix was prescribed a series of loading, material, and architectural parameters. The distribution of CME in the scaffold was predicted and compared to proposed target mechanics based on anabolic responses of AF cells. RESULTS The multi-axial loading modality predicted the greatest percentage of cell volumes falling within the CME target envelope (%PTE) in the study (65 %PTE for 5.0% equibiaxial tensile strain with 50 kPa radial-direction compression; 7.6 %PTE without radial pressure). Additionally, the architectural scale had a moderate influence on the CME (maximum of 17 %PTE), with minimal change in the tissue-level properties of the scaffold. Scaffold materials and architectures had secondary influences on the predicted regeneration by modifying the tissue-level scaffold mechanics. CONCLUSIONS Scaffold loading modality was identified as the critical factor for TE the AF. Scaffold materials and architecture were also predicted to modulate the scaffold loading and, therefore, control the CME indirectly. This study facilitated an improved understanding of the relationship between tissue-level and cell-level mechanics to drive anabolic cell responses for tissue regeneration.
Collapse
Affiliation(s)
- Mitchell I. Page
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering and School of Biomedical EngineeringColorado State UniversityFt CollinsColoradoUSA
| | - Peter E. Linde
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering and School of Biomedical EngineeringColorado State UniversityFt CollinsColoradoUSA
| | - Christian M. Puttlitz
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering and School of Biomedical EngineeringColorado State UniversityFt CollinsColoradoUSA
| |
Collapse
|
16
|
Ke S, Liu Q, Zhang X, Yao Y, Yang X, Sui G. Cytotoxicity analysis of biomass combustion particles in human pulmonary alveolar epithelial cells on an air-liquid interface/dynamic culture platform. Part Fibre Toxicol 2021; 18:31. [PMID: 34419099 PMCID: PMC8379799 DOI: 10.1186/s12989-021-00426-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/05/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Exposure to indoor air pollution from solid fuel combustion is associated with lung diseases and cancer. This study investigated the cytotoxicity and molecular mechanisms of biomass combustion-derived particles in human pulmonary alveolar epithelial cells (HPAEpiC) using a platform that combines air-liquid interface (ALI) and dynamic culture (DC) systems. METHODS HPAEpiC were cultured on the surface of polycarbonate (PC) membranes on the ALI-DC platform. The cells were sprayed with an aerosolized solution of biomass combustion soluble constituents (BCSCs) and simultaneously nourished with culture medium flowing beneath the permeable PC membranes. The ALI-DC method was compared with the traditional submerged culture approach. BCSC particle morphology and dosages deposited on the chip were determined for particle characterization. Flow cytometry, scanning electron microscopy, and transmission electron microscopy were used to investigate the apoptosis rate of HPAEpiC and changes in the cell ultrastructure induced by BCSCs. Additionally, the underlying apoptotic pathway was examined by determining the protein expression levels by western blotting. RESULTS Scanning electron microscope images demonstrated that the sample processing and delivering approach of the ALI-DC platform were suitable for pollutant exposure. Compared with the submerged culture method, a significant decline in cell viability and increase in apoptosis rate was observed after BCSC exposure on the ALI-DC platform, indicating that the ALI-DC platform is a more sensitive system for investigating cytotoxicity of indoor air pollutants in lung cells. The morphology and ultrastructure of the cells were damaged after exposure to BCSCs, and the p53 pathway was activated. The Bcl-2/Bax ratio was reduced, upregulating caspase-9 and caspase-3 expression and subsequently inducing apoptosis of HPAEpiC. The addition of N-acetyl cysteine antioxidant significantly alleviated the cytotoxicity induced by BCSCs. CONCLUSION A novel ALI-DC platform was developed to study the cytotoxicity of air pollutants on lung cells. Using the platform, we demonstrated that BCSCs could damage the mitochondria, produce reactive oxygen species, and activate p53 in HPAEpiC, ultimately inducing apoptosis.
Collapse
Affiliation(s)
- Shaorui Ke
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046 People’s Republic of China
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai, 200433 People’s Republic of China
| | - Qi Liu
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai, 200433 People’s Republic of China
| | - Xinlian Zhang
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai, 200433 People’s Republic of China
| | - Yuhan Yao
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai, 200433 People’s Republic of China
| | - Xudong Yang
- Department of Building Science, Tsinghua University, Beijing, 100084 People’s Republic of China
| | - Guodong Sui
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai, 200433 People’s Republic of China
- Jiangsu Collaborative Innovation Center of Atmospheric Environment and Equipment Technology (CICAEET), Nanjing University of Information Science & Technology, Nanjing, 210044 People’s Republic of China
| |
Collapse
|
17
|
Peredo AP, Gullbrand SE, Smith HE, Mauck RL. Putting the Pieces in Place: Mobilizing Cellular Players to Improve Annulus Fibrosus Repair. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:295-312. [PMID: 32907498 PMCID: PMC10799291 DOI: 10.1089/ten.teb.2020.0196] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The intervertebral disc (IVD) is an integral load-bearing tissue that derives its function from its composite structure and extracellular matrix composition. IVD herniations involve the failure of the annulus fibrosus (AF) and the extrusion of the nucleus pulposus beyond the disc boundary. Disc herniations can impinge the neural elements and cause debilitating pain and loss of function, posing a significant burden on individual patients and society as a whole. Patients with persistent symptoms may require surgery; however, surgical intervention fails to repair the ruptured AF and is associated with the risk for reherniation and further disc degeneration. Given the limitations of AF endogenous repair, many attempts have been made toward the development of effective repair approaches that reestablish IVD function. These methods, however, fail to recapitulate the composition and organization of the native AF, ultimately resulting in inferior tissue mechanics and function over time and high rates of reherniation. Harnessing the cellular function of cells (endogenous or exogenous) at the repair site through the provision of cell-instructive cues could enhance AF tissue regeneration and, ultimately, improve healing outcomes. In this study, we review the diverse approaches that have been developed for AF repair and emphasize the potential for mobilizing the appropriate cellular players at the site of injury to improve AF healing. Impact statement Conventional treatments for intervertebral disc herniation fail to repair the annulus fibrosus (AF), increasing the risk for recurrent herniation. The lack of repair devices in the market has spurred the development of regenerative approaches, yet most of these rely on a scarce endogenous cell population to repair large injuries, resulting in inadequate regeneration. This review identifies current and developing strategies for AF repair and highlights the potential for harnessing cellular function to improve AF regeneration. Ideal cell sources, differentiation strategies, and delivery methods are discussed to guide the design of repair systems that leverage specialized cells to achieve superior outcomes.
Collapse
Affiliation(s)
- Ana P. Peredo
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Sarah E. Gullbrand
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Harvey E. Smith
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Robert L. Mauck
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Affiliation(s)
- Kanchan Maji
- Center of Excellence in Tissue Engineering, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, India
| | - Krishna Pramanik
- Center of Excellence in Tissue Engineering, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, India
| |
Collapse
|
19
|
Smoak MM, Hogan KJ, Grande-Allen KJ, Mikos AG. Bioinspired electrospun dECM scaffolds guide cell growth and control the formation of myotubes. SCIENCE ADVANCES 2021; 7:eabg4123. [PMID: 33990336 PMCID: PMC8121426 DOI: 10.1126/sciadv.abg4123] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/25/2021] [Indexed: 06/01/2023]
Abstract
While skeletal muscle has a high capacity for endogenous repair in acute injuries, volumetric muscle loss can leave long-lasting or permanent structural and functional deficits to the injured muscle and surrounding tissues. With clinical treatments failing to repair lost tissue, there is a great need for a tissue-engineered therapy to promote skeletal muscle regeneration. In this study, we aim to assess the potential for electrospun decellularized skeletal muscle extracellular matrix (dECM) with tunable physicochemical properties to control mouse myoblast growth and myotube formation. The material properties as well as cell behavior - growth and differentiation - were assessed in response to modulation of crosslinking and scaffold architecture. The fabrication of a bioactive dECM-based system with tunable physicochemical properties that can control myotube formation has several applications in skeletal muscle engineering and may bring the field one step closer to developing a therapy to address these unmet clinical needs.
Collapse
Affiliation(s)
- Mollie M Smoak
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Katie J Hogan
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | | | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
20
|
Locke RC, Ford EM, Silbernagel KG, Kloxin AM, Killian ML. Success Criteria and Preclinical Testing of Multifunctional Hydrogels for Tendon Regeneration. Tissue Eng Part C Methods 2020; 26:506-518. [PMID: 32988293 PMCID: PMC7869878 DOI: 10.1089/ten.tec.2020.0199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/20/2020] [Indexed: 12/16/2022] Open
Abstract
Tendon injuries are difficult to heal, in part, because intrinsic tendon healing, which is dominated by scar tissue formation, does not effectively regenerate the native structure and function of healthy tendon. Further, many current treatment strategies also fall short of producing regenerated tendon with the native properties of healthy tendon. There is increasing interest in the use of cell-instructive strategies to limit the intrinsic fibrotic response following injury and improve the regenerative capacity of tendon in vivo. We have established multifunctional, cell-instructive hydrogels for treating injured tendon that afford tunable control over the biomechanical, biochemical, and structural properties of the cell microenvironment. Specifically, we incorporated integrin-binding domains (RGDS) and assembled multifunctional collagen mimetic peptides that enable cell adhesion and elongation of stem cells within synthetic hydrogels of designed biomechanical properties and evaluated these materials using targeted success criteria developed for testing in mechanically demanding environments such as tendon healing. The in vitro and in situ success criteria were determined based on systematic reviews of the most commonly reported outcome measures of hydrogels for tendon repair and established standards for testing of biomaterials. We then showed, using validation experiments, that multifunctional and synthetic hydrogels meet these criteria. Specifically, these hydrogels have mechanical properties comparable to developing tendon; are noncytotoxic both in two-dimensional bolus exposure (hydrogel components) and three-dimensional encapsulation (full hydrogel); are formed, retained, and visualized within tendon defects over time (2-weeks); and provide mechanical support to tendon defects at the time of in situ gel crosslinking. Ultimately, the in vitro and in situ success criteria evaluated in this study were designed for preclinical research to rigorously test the potential to achieve successful tendon repair before in vivo testing and indicate the promise of multifunctional and synthetic hydrogels for continued translation. Impact statement Tendon healing results in a weak scar that forms due to poor cell-mediated repair of the injured tissue. Treatments that tailor the instructions experienced by cells during healing afford opportunities to regenerate the healthy tendon. Engineered cell-instructive cues, including the biomechanical, biochemical, and structural properties of the cell microenvironment, within multifunctional synthetic hydrogels are promising therapeutic strategies for tissue regeneration. In this article, the preclinical efficacy of multifunctional synthetic hydrogels for tendon repair is tested against rigorous in vitro and in situ success criteria. This study indicates the promise for continued preclinical translation of synthetic hydrogels for tissue regeneration.
Collapse
Affiliation(s)
- Ryan C. Locke
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| | - Eden M. Ford
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | | | - April M. Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
| | - Megan L. Killian
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
21
|
Turner PR, Yoshida M, Ali MA, Cabral JD. Melt Electrowritten Sandwich Scaffold Technique Using Sulforhodamine B to Monitor Stem Cell Behavior. Tissue Eng Part C Methods 2020; 26:519-527. [PMID: 32977739 DOI: 10.1089/ten.tec.2020.0240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Three-dimensional (3D) printing using melt electrowriting (MEW) technology is a recently developed technique to produce biocompatible micron-level mesh scaffolds layer-by-layer that can be seeded with cells for tissue engineering. Examining cell behavior, such as growth rate and migration, can be problematic in these opaque 3D scaffolds. A straightforward and quantitative method was developed to examine these cellular parameters on poly-ɛ-caprolactone (PCL) multilayered MEW scaffolds developed as components of the annulus fibrosus region of bioengineered intervertebral discs. Experiment: The anti-adhesion protein, bovine serum albumin (BSA), was used to coat plasticware to improve mesenchymal stem cell (T0523) adhesion to MEW scaffolds. Cells were seeded on circular MEW (cMEW) discs as defined growth starting points sandwiched between two test template scaffolds investigated at varying pore sizes. Cell expansion, growth, and migration were quantitated utilizing the protein-specific dye sulforhodamine B (SRB). Live cell imaging combined with image analysis were used to examine cell motility and expansion on 3D scaffolds. Results: After one coating of BSA, cells remained nonadherent for the duration of the study with cell spheroids formed and enlarging over 21 days and becoming entangled in MEW scaffold pores. Cells grown on the 250 μm pore size scaffolds exhibited a doubling time of 7 days, whereas the 400 μm pore size scaffolds time was 11.5 days. Conclusions: BSA coating of tissue culture dishes prevented surface adhesion of cells to vessel surfaces and promoted spheroid formation that encouraged attachment to the PCL scaffolds. Batch-printed cMEW scaffolds were useful as a defined starting point for quantitative assays that successfully measured cell migration, expansion and proliferation on test scaffolds. The SRB assay was shown to be a useful and straightforward way to quantitate cell numbers in multilayered MEW scaffolds. A pore size of 250 μm exhibited the fastest cell growth, spread, and expansion. Impact statement In this article, a new, useful, and straightforward method to quantitate cell numbers on three-dimensional (3D) melt electrowritten (MEW) scaffolds is presented. By using the sulforhodamine B assay on bovine serum albumin-coated dishes cell migration, expansion and proliferation in 3D printed MEW test scaffolds were quantitatively measured. Printed circular MEW (cMEW) scaffolds sandwiched between two MEW test scaffolds (Fig. 3) were used as defined cellular growth starting points with a particular pore size of 250 μm displaying the fastest cell growth and migration. This MEW sandwich technique could potentially be used to quantitate cell numbers and migration in other 3D multilayered MEW scaffold systems.
Collapse
Affiliation(s)
- Paul R Turner
- Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - Minami Yoshida
- Department of Food Science, Centre of Bioengineering & Nanomedicine, University of Otago, Dunedin, New Zealand
| | - M Azam Ali
- Department of Food Science, Centre of Bioengineering & Nanomedicine, University of Otago, Dunedin, New Zealand
| | - Jaydee D Cabral
- Department of Chemistry, University of Otago, Dunedin, New Zealand.,Department of Food Science, Centre of Bioengineering & Nanomedicine, University of Otago, Dunedin, New Zealand
| |
Collapse
|
22
|
Orkwis JA, Wolf AK, Shahid SM, Smith C, Esfandiari L, Harris GM. Development of a Piezoelectric PVDF-TrFE Fibrous Scaffold to Guide Cell Adhesion, Proliferation, and Alignment. Macromol Biosci 2020; 20:e2000197. [PMID: 32691517 DOI: 10.1002/mabi.202000197] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/28/2020] [Indexed: 12/20/2022]
Abstract
Severe peripheral nervous system injuries currently hold limited therapeutic solutions. Existing clinical techniques such as autografts, allografts, and newer nerve guidance conduits have shown variable outcomes in functional recovery, adverse immune responses, and in some cases low or minimal availability. This can be attributed in part to the lack of chemical, physical, and electrical cues directing both nerve guidance and regeneration. To address this pressing clinical issue, electrospun nanofibers and microfibers composed of piezoelectric polyvinylidene flouride-triflouroethylene (PVDF-TrFE) have been introduced as an alternative template for tissue engineered biomaterials, specifically as it pertains to their relevance in soft tissue and nerve repair. Here, biocompatible scaffolds of PVDF-TrFE are fabricated and their ability to generate an electrical response to mechanical deformations and produce a suitable regenerative microenvironment is examined. It is determined that 20% (w/v) PVDF-TrFE in (6:4) dimethyl formamide (DMF):acetone solvent maintains a desirable piezoelectric coefficient and the proper physical and electrical characteristics for tissue regeneration. Further, it is concluded that scaffolds of varying thickness promoted the adhesion and alignment of Schwann cells and fibroblasts. This work offers a prelude to further advancements in nanofibrous technology and a promising outlook for alternative, autologous remedies to peripheral nerve damage.
Collapse
Affiliation(s)
- Jacob A Orkwis
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Ann K Wolf
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Syed M Shahid
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Corinne Smith
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Leyla Esfandiari
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, 45221, USA.,Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Greg M Harris
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA.,Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA.,Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| |
Collapse
|
23
|
Panebianco C, DiStefano T, Mui B, Hom W, Iatridis J. Crosslinker concentration controls TGFβ-3 release and annulus fibrosus cell apoptosis in genipin-crosslinked fibrin hydrogels. Eur Cell Mater 2020; 39:211-226. [PMID: 32396210 PMCID: PMC7372750 DOI: 10.22203/ecm.v039a14] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Back pain is a leading cause of global disability associated with intervertebral disc (IVD) pathologies. Discectomy alleviates disabling pain caused by IVD herniation without repairing annulus fibrosus (AF) defects, which can cause accelerated degeneration and recurrent pain. Biological therapies show promise for IVD repair but developing high-modulus biomaterials capable of providing biomechanical stabilisation and delivering biologics remains an unmet challenge. The present study identified critical factors and developed an optimal formulation to enhance the delivery of AF cells and transforming growth beta-3 (TGFβ-3) in genipin-crosslinked fibrin (FibGen) hydrogels. Part 1 showed that AF cells encapsulated in TGFβ-3-supplemented high-modulus FibGen synthesised little extracellular matrix (ECM) but could release TGFβ-3 at physiologically relevant levels. Part 2 showed that AF cells underwent apoptosis when encapsulated in FibGen, even after reducing fibrin concentration from 70 to 5 mg/mL. Mechanistic experiments, modifying genipin concentration and integrin binding site presence demonstrated that genipin crosslinking caused AF cell apoptosis by inhibiting cell-biomaterial binding. Adding integrin binding sites with fibronectin partially rescued apoptosis, indicating genipin also caused acute cytotoxicity. Part 3 showed that FibGen formulations with 1 mg/mL genipin had enhanced ECM synthesis when supplemented with fibronectin and TGFβ-3. In conclusion, FibGen could be used for delivering biologically active compounds and AF cells, provided that formulations supplied additional sites for cell-biomaterial binding and genipin concentrations were low. Results also highlighted a need for developing strategies that protect cells against acute crosslinker cytotoxicity to overcome challenges of engineering high-modulus cell carriers for musculoskeletal tissues that experience high mechanical demands.
Collapse
Affiliation(s)
| | | | | | | | - J.C. Iatridis
- Address for correspondence: James C. Iatridis, PhD, 1468 Madison Avenue-Annenberg Building, floor 20, Room A20-086, New York, 10029 NY, USA., Telephone number: +1 2122411517
| |
Collapse
|
24
|
Zhao H, Tang J, Zhou D, Weng Y, Qin W, Liu C, Lv S, Wang W, Zhao X. Electrospun Icariin-Loaded Core-Shell Collagen, Polycaprolactone, Hydroxyapatite Composite Scaffolds for the Repair of Rabbit Tibia Bone Defects. Int J Nanomedicine 2020; 15:3039-3056. [PMID: 32431500 PMCID: PMC7200251 DOI: 10.2147/ijn.s238800] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Background Electrospinning is a widely used technology that can produce scaffolds with high porosity and surface area for bone regeneration. However, the small pore sizes in electrospun scaffolds constrain cell growth and tissue-ingrowth. In this study, novel drug-loading core-shell scaffolds were fabricated via electrospinning and freeze drying to facilitate the repair of tibia bone defects in rabbit models. Materials and Methods The collagen core scaffolds were freeze-dried containing icariin (ICA)-loaded chitosan microspheres. The shell scaffolds were electrospun using collagen, polycaprolactone and hydroxyapatite materials to form CPH composite scaffolds with the ones containing ICA microspheres named CPHI. The core-shell scaffolds were then cross-linked by genipin. The morphology, microstructure, physical and mechanical properties of the scaffolds were assessed. Rat marrow mesenchymal stem cells from the wistar rat were cultured with the scaffolds. The cell adhesion and proliferation were analysed. Adult rabbit models with tibial plateau defects were used to evaluate the performance of these scaffolds in repairing the bone defects over 4 to 12 weeks. Results The results reveal that the novel drug-loading core-shell scaffolds were successfully fabricated, which showed good physical and chemical properties and appropriate mechanical properties. Furthermore, excellent cells attachment was observed on the CPHI scaffolds. The results from radiography, micro-computed tomography, histological and immunohistochemical analysis demonstrated that abundant new bones were formed on the CPHI scaffolds. Conclusion These new core-shell composite scaffolds have great potential for bone tissue engineering applications and may lead to effective bone regeneration and repair.
Collapse
Affiliation(s)
- Hongbin Zhao
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Junjie Tang
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Dong Zhou
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Yiping Weng
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Wen Qin
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Chun Liu
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Songwei Lv
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, People's Republic of China
| | - Wei Wang
- Medical School, Hexi University, Zhangye 730041, People's Republic of China
| | - Xiubo Zhao
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, People's Republic of China.,Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| |
Collapse
|
25
|
Bongiovanni Abel S, Montini Ballarin F, Abraham GA. Combination of electrospinning with other techniques for the fabrication of 3D polymeric and composite nanofibrous scaffolds with improved cellular interactions. NANOTECHNOLOGY 2020; 31:172002. [PMID: 31931493 DOI: 10.1088/1361-6528/ab6ab4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The development of three-dimensional (3D) scaffolds with physical and chemical topological cues at the macro-, micro-, and nanometer scale is urgently needed for successful tissue engineering applications. 3D scaffolds can be manufactured by a wide variety of techniques. Electrospinning technology has emerged as a powerful manufacturing technique to produce non-woven nanofibrous scaffolds with very interesting features for tissue engineering products. However, electrospun scaffolds have some inherent limitations that compromise the regeneration of thick and complex tissues. By integrating electrospinning and other fabrication technologies, multifunctional 3D fibrous assemblies with micro/nanotopographical features can be created. The proper combination of techniques leads to materials with nano and macro-structure, allowing an improvement in the biological performance of tissue-engineered constructs. In this review, we focus on the most relevant strategies to produce electrospun polymer/composite scaffolds with 3D architecture. A detailed description of procedures involving physical and chemical agents to create structures with large pores and 3D fiber assemblies is introduced. Finally, characterization and biological assays including in vitro and in vivo studies of structures intended for the regeneration of functional tissues are briefly presented and discussed.
Collapse
Affiliation(s)
- Silvestre Bongiovanni Abel
- Research Institute for Materials Science and Technology, INTEMA (UNMdP-CONICET). Av. Colón 10850, B7606BWV, Mar del Plata, Argentina
| | | | | |
Collapse
|
26
|
Theodoridis K, Aggelidou E, Manthou M, Demiri E, Bakopoulou A, Kritis A. Assessment of cartilage regeneration on 3D collagen-polycaprolactone scaffolds: Evaluation of growth media in static and in perfusion bioreactor dynamic culture. Colloids Surf B Biointerfaces 2019; 183:110403. [PMID: 31400614 DOI: 10.1016/j.colsurfb.2019.110403] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 07/23/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022]
Abstract
Efforts on bioengineering are directed towards the construction of biocompatible scaffolds and the determination of the most favorable microenvironment, which will better support cell proliferation and differentiation. Perfusion bioreactors are attracting growing attention as an effective, modern tool in tissue engineering. A natural biomaterial extensively used in regenerative medicine with outstanding biocompatibility, biodegradability and non-toxic characteristics, is collagen, a structural protein with undisputed beneficial characteristics. This is a study designed according to the above considerations. 3D printed polycaprolactone (PCL) scaffolds with rectangular pores were coated with collagen either as a coating on the scaffold's trabeculae, or as a gel-cell solution penetrating scaffolds' pores. We employed histological, molecular and imaging techniques to analyze colonization, proliferation and chondrogenic differentiation of Adipose Derived Mesenchymal Stem Cells (ADMSCs). Two different differentiation culture media were employed to test chondrogenic differentiation on gelated and non gelated PCL scaffolds in static and in perfusion bioreactors dynamic culture conditions. In dynamic culture, non gelated scaffolds combined with our in house TGF-β2 based medium, augmented chondrogenic differentiation performance, which overall was significantly less favorable compared to StemPro™ propriety medium. The beneficial mechanical stimulus of dynamic culture, appears to outgrow the disadvantage of the "weaker" TGF-β2 medium used for chondrogenic differentiation. Even though cells in static culture grew well on the scaffold, there was limited penetration inside the construct, so the purpose of the 3D culture was not fully served. In contrast dynamic culture achieved better penetration and uniform distribution of the cells within the scaffold.
Collapse
Affiliation(s)
- Konstantinos Theodoridis
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece; cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Eleni Aggelidou
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece; cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Marilena Manthou
- cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece; Laboratory of Histology, Embryology and Anthropology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Efterpi Demiri
- Department of Plastic Surgery, Medical School, Papageorgiou Hospital, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Athina Bakopoulou
- cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece; Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), GR-54124, Thessaloniki, Greece
| | - Aristeidis Kritis
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece; cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece.
| |
Collapse
|
27
|
Zhao R, Liu W, Xia T, Yang L. Disordered Mechanical Stress and Tissue Engineering Therapies in Intervertebral Disc Degeneration. Polymers (Basel) 2019; 11:polym11071151. [PMID: 31284436 PMCID: PMC6680713 DOI: 10.3390/polym11071151] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 12/11/2022] Open
Abstract
Low back pain (LBP), commonly induced by intervertebral disc degeneration, is a lumbar disease with worldwide prevalence. However, the mechanism of degeneration remains unclear. The intervertebral disc is a nonvascular organ consisting of three components: Nucleus pulposus, annulus fibrosus, and endplate cartilages. The disc is structured to support our body motion and endure persistent external mechanical pressure. Thus, there is a close connection between force and intervertebral discs in LBP. It is well established that with aging, disordered mechanical stress profoundly influences the fate of nucleus pulposus and the alignment of collagen fibers in the annulus fibrosus. These support a new understanding that disordered mechanical stress plays an important role in the degeneration of the intervertebral discs. Tissue-engineered regenerative and reparative therapies are being developed for relieving disc degeneration and symptoms of lower back pain. In this paper, we will review the current literature available on the role of disordered mechanical stress in intervertebral disc degeneration, and evaluate the existing tissue engineering treatment strategies of the current therapies.
Collapse
Affiliation(s)
- Runze Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Wanqian Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Tingting Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
28
|
Wang W, Wan Y, Fu T, Zhou T, Tang X, Wu H, Liu C, Jagodzinski M. Effect of cyclic compression on bone marrow mesenchymal stromal cells in tissue engineered cartilage scaffold. J Biomed Mater Res A 2019; 107:1294-1302. [PMID: 30707490 DOI: 10.1002/jbm.a.36642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/11/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022]
Abstract
In this current study, a novel multilayer porous composite scaffold was fabricated with chitosan (CS), silk fibrin (SF) and nano-hydroxyapatite (n-HA). Scanning electron microscope was utilized to detect the characteristics of the composed scaffold. Rat bone marrow stromal cells (rBMSC) were loaded onto the CS/SF/n-HA scaffold and cultured in a bioreactor under an on-off dynamic compression (10% compressive strain, 0.5 Hz, [2 h action + 4 h pause]/cycle, 4 cycles/day). Metabolism of the loaded rBMSC was assessed through CCK-8 test. Qualitative polymerase chain reaction and western blot were applied to assess the chondrogenic differentiation of the seeded cells. Compressive modulus of the cell/scaffold constructs was analyzed. Additionally, a pig model was employed to evaluate the effect of the tissue-engineered cartilage on repairing of cartilage defect. Results showed that the four layers within the scaffold were tightly connected without gaps between porous interfaces of the layers. Scaffold porosity was 92.20% ± 1.30%. The cyclic compression upregulated chondrogenesis markers (Aggrecan, Sox-9, and collagen II). Increased compressive modulus of the cell/scaffold complex was detected after dynamic compression. The pig bone marrow stromal cells/scaffold complex exposed to cyclic compression presented most favorable reparative effect on the mini pig femoral condyle cartilage defects. Our study suggested that the on-off dynamic compression might be a promising approach to fabricate tissue-engineered cartilage in vitro. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1294-1302, 2019.
Collapse
Affiliation(s)
- Wei Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Wan
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tao Fu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Zhou
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangyu Tang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chaoxu Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Michael Jagodzinski
- Department of Orthopedic Trauma, Hanover Medical School (MHH), Hanover D-30625, Germany
| |
Collapse
|
29
|
Chen M, Guo W, Gao S, Hao C, Shen S, Zhang Z, Wang Z, Li X, Jing X, Zhang X, Yuan Z, Wang M, Zhang Y, Peng J, Wang A, Wang Y, Sui X, Liu S, Guo Q. Biomechanical Stimulus Based Strategies for Meniscus Tissue Engineering and Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:392-402. [PMID: 29897012 DOI: 10.1089/ten.teb.2017.0508] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Meniscus injuries are very common in the knee joint. Treating a damaged meniscus continues to be a scientific challenge in sport medicine because of its poor self-healing potential and few clinical therapeutic options. Tissue engineering strategies are very promising solutions for repairing and regenerating a damaged meniscus. Meniscus is exposed to a complex biomechanical microenvironment, and it plays a crucial role in meniscal development, growth, and repairing. Over the past decades, increasing attention has been focused on the use of biomechanical stimulus to enhance biomechanical properties of the engineered meniscus. Further understanding the influence of mechanical stimulation on cell proliferation and differentiation, metabolism, relevant gene expression, and pro/anti-inflammatory responses may be beneficial to enhance meniscal repair and regeneration. On the one hand, this review describes some basic information about meniscus; on the other hand, we sum up the various biomechanical stimulus based strategies applied in meniscus tissue engineering and how these factors affect meniscal regeneration. We hope this review will provide researchers with inspiration on tissue engineering strategies for meniscus regeneration in the future.
Collapse
Affiliation(s)
- Mingxue Chen
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China .,2 Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, 100035 Beijing, People's Republic of China
| | - Weimin Guo
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Shunag Gao
- 3 Center for Biomaterial and Tissue Engineering, Academy for Advanced Interdisciplinary Studies, Peking University , Beijing, People's Republic of China
| | - Chunxiang Hao
- 4 Institute of Anesthesiology , Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Shi Shen
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China .,5 Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University , Luzhou, People's Republic of China
| | - Zengzeng Zhang
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China .,6 First Department of Orthopedics, First Affiliated Hospital of Jiamusi University , Jiamusi, People's Republic of China
| | - Zehao Wang
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Xu Li
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China .,7 School of Medicine, Nankai University , Tianjin, People's Republic of China
| | - Xiaoguang Jing
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China .,6 First Department of Orthopedics, First Affiliated Hospital of Jiamusi University , Jiamusi, People's Republic of China
| | - Xueliang Zhang
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China .,8 Shanxi Traditional Chinese Hospital , Taiyuan, People's Republic of China
| | - Zhiguo Yuan
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Mingjie Wang
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Yu Zhang
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Jiang Peng
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Aiyuan Wang
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Yu Wang
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Xiang Sui
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Shuyun Liu
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| | - Quanyi Guo
- 1 Institute of Orthopedics , Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, People's Republic of China
| |
Collapse
|
30
|
D'Amore A, Nasello G, Luketich SK, Denisenko D, Jacobs DL, Hoff R, Gibson G, Bruno A, T Raimondi M, Wagner WR. Meso-scale topological cues influence extracellular matrix production in a large deformation, elastomeric scaffold model. SOFT MATTER 2018; 14:8483-8495. [PMID: 30357253 DOI: 10.1039/c8sm01352g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Physical cues are decisive factors in extracellular matrix (ECM) formation and elaboration. Their transduction across scale lengths is an inherently symbiotic phenomenon that while influencing ECM fate is also mediated by the ECM structure itself. This study investigates the possibility of enhancing ECM elaboration by topological cues that, while not modifying the substrate macro scale mechanics, can affect the meso-scale strain range acting on cells incorporated within the scaffold. Vascular smooth muscle cell micro-integrated, electrospun scaffolds were fabricated with comparable macroscopic biaxial mechanical response, but different meso-scale topology. Seeded scaffolds were conditioned on a stretch bioreactor and exposed to large strain deformations. Samples were processed to evaluate ECM quantity and quality via: biochemical assay, qualitative and quantitative histological assessment and multi-photon analysis. Experimental evaluation was coupled to a numerical model that elucidated the relationship between the scaffold micro-architecture and the strain acting on the cells. Results showed an higher amount of ECM formation for the scaffold type characterized by lowest fiber intersection density. The numerical model simulations associated this result with the differences found for the change in cell nuclear aspect ratio and showed that given comparable macro scale mechanics, a difference in material topology created significant differences in cell-scaffold meso-scale deformations. These findings reaffirmed the role of cell shape in ECM formation and introduced a novel notion for the engineering of cardiac tissue where biomaterial structure can be designed to both mimick the organ level mechanics of a specific tissue of interest and elicit a desirable cellular response.
Collapse
Affiliation(s)
- Antonio D'Amore
- Departments of Bioengineering and Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, 15216, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ke S, Liu Q, Deng M, Zhang X, Yao Y, Shan M, Yang X, Sui G. Cytotoxicity analysis of indoor air pollution from biomass combustion in human keratinocytes on a multilayered dynamic cell culture platform. CHEMOSPHERE 2018; 208:1008-1017. [PMID: 30068025 DOI: 10.1016/j.chemosphere.2018.06.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/07/2018] [Indexed: 06/08/2023]
Abstract
Skin tissue is the first barrier against ambient harmful matter and has direct contact with indoor air pollutants. Nevertheless, a comprehensive understanding of cytotoxicity of indoor air pollution on skin cells is insufficiently clear. Herein, for the first time a multilayered dynamic cell culture platform was established to study the cytotoxicity of indoor air pollutant from biomass combustion in human skin keratinocytes. The platform consisted of seven repetitive polydimethylsiloxane modules carrying six pieces of polycarbonate membrane between them as substrate for cell growth to realize the simultaneous dynamic culture of 12 layers of keratinocytes. After exposure to biomass combustion soluble constituents (BCSCs), cell viability under microfluidic platform conditions declined more significantly, and apoptosis rates increased more obviously compared with well plate conditions. Transmission electron microscope showed that keratinocyte microstructures displayed obvious signs of cellular damage. Our study confirmed that the nuclear factor of kappa B (NF-κB) signaling pathway was activated, which significantly increased the Bax/Bcl-2 ratio and tumor necrosis factor-alpha and interleukin 6 expression, indicating that NF-κB signaling pathway was the major factor in BCSCs-induced cytotoxicity. These findings offer an insight into the mechanism of BCSCs-induced cytotoxicity in keratinocytes and provide a theoretical basis for future studies on skin cells.
Collapse
Affiliation(s)
- Shaorui Ke
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai 200433, PR China
| | - Qi Liu
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai 200433, PR China
| | - Mengsi Deng
- Department of Building Science, Tsinghua University, Beijing 100084, PR China
| | - Xinlian Zhang
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai 200433, PR China
| | - Yuhan Yao
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai 200433, PR China
| | - Ming Shan
- Department of Building Science, Tsinghua University, Beijing 100084, PR China
| | - Xudong Yang
- Department of Building Science, Tsinghua University, Beijing 100084, PR China.
| | - Guodong Sui
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai 200433, PR China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, PR China.
| |
Collapse
|
32
|
Chu G, Shi C, Wang H, Zhang W, Yang H, Li B. Strategies for Annulus Fibrosus Regeneration: From Biological Therapies to Tissue Engineering. Front Bioeng Biotechnol 2018; 6:90. [PMID: 30042942 PMCID: PMC6048238 DOI: 10.3389/fbioe.2018.00090] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/18/2018] [Indexed: 12/17/2022] Open
Abstract
Intervertebral disc (IVD) is an avascular tissue which contributes to the weight bearing, motion, and flexibility of spine. However, IVD is susceptible to damage and even failure due to injury, pathology, and aging. Annulus fibrosus (AF), the structural and functional integrity of which is critically essential to confine nucleus pulpous (NP) and maintain physiological intradiscal pressure under mechanical loading, plays a critical role in the biomechanical properties of IVD. AF degeneration commonly results in substantial deterioration of IVD. During this process, the biomechanical properties of AF and the balance between anabolism and catabolism in IVD are progressively disrupted, leading to chronic back pain, and even disability of individuals. Therefore, repairing and regenerating AF are effective treatments to degeneration-associated pains. However, they remain highly challenging due to the complexity of natural AF tissue in the aspects of cell phenotype, biochemical composition, microstructure, and mechanical properties. Tissue engineering (TE), by combining biological science and materials engineering, shed lights on AF regeneration. In this article, we review recent advances in the pro-anabolic approaches in the form of cell delivery, bioactive factors delivery, gene therapy, and TE strategies for achieving AF regeneration.
Collapse
Affiliation(s)
- Genglei Chu
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedic Surgery, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Chen Shi
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Huan Wang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedic Surgery, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Weidong Zhang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedic Surgery, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Huilin Yang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedic Surgery, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Bin Li
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedic Surgery, First Affiliated Hospital, Soochow University, Suzhou, China
- China Orthopaedic Regenerative Medicine Group, Hangzhou, China
| |
Collapse
|
33
|
Fan L, Li JL, Cai Z, Wang X. Creating Biomimetic Anisotropic Architectures with Co-Aligned Nanofibers and Macrochannels by Manipulating Ice Crystallization. ACS NANO 2018; 12:5780-5790. [PMID: 29846058 DOI: 10.1021/acsnano.8b01648] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The continuous evolution of tissue engineering scaffolds has been driven by the desire to recapitulate structural features and functions of the natural extracellular matrix (ECM). However, it is still an extreme challenge to create a three-dimensional (3D) scaffold with both aligned nanofibers and aligned interconnected macrochannels to mimic the ECM of anisotropic tissues. Here, we develop a facile strategy to create such a scaffold composed of oriented nanofibers and interconnected macrochannels in the same direction, with various natural polymers typically used for tissue regeneration. The orientation of nanofibers and interconnected macrochannels can be easily tuned by manipulating ice crystallization. The scaffold demonstrates both structural and functional features similar to the natural ECM of anisotropic tissues. Taking silk fibroin as an example, the scaffold with radially oriented nanofibers and interconnected macrochannels is more efficient for capturing cells and promoting the growth of both nonadherent embryonic dorsal root ganglion neurons (DRGs) and adherent human umbilical vein endothelial cells (HUVECs) compared to the widely used scaffold types. Interestingly, DRGs and neurites on the SF scaffold demonstrate a 3D growth mode similar to that of natural nerve tissues. Furthermore, the coaligned nanofibers and macrochannels of the scaffold can direct HUVECs to assemble into blood vessel-like structures and their collagen deposition in their arrangement direction. The strategy could inspire the design and development of multifunctional 3D scaffolds with desirable structural features for engineering different tissues.
Collapse
Affiliation(s)
- Linpeng Fan
- Institute for Frontier Materials , Deakin University , Geelong , Victoria 3216 , Australia
| | - Jing-Liang Li
- Institute for Frontier Materials , Deakin University , Geelong , Victoria 3216 , Australia
| | - Zengxiao Cai
- Institute for Frontier Materials , Deakin University , Geelong , Victoria 3216 , Australia
| | - Xungai Wang
- Institute for Frontier Materials , Deakin University , Geelong , Victoria 3216 , Australia
| |
Collapse
|
34
|
Ma J, He Y, Liu X, Chen W, Wang A, Lin CY, Mo X, Ye X. A novel electrospun-aligned nanoyarn/three-dimensional porous nanofibrous hybrid scaffold for annulus fibrosus tissue engineering. Int J Nanomedicine 2018; 13:1553-1567. [PMID: 29588584 PMCID: PMC5858820 DOI: 10.2147/ijn.s143990] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Introduction Herniation of the nucleus pulposus (NP) because of defects in the annulus fibrosus (AF) is a well-known cause of low back pain. Defects in the AF thus remain a surgical challenge, and efforts have been made to develop new techniques for closure and repair. In this study, we developed an electrospun aligned nanoyarn scaffold (AYS) and nanoyarn/three-dimensional porous nanofibrous hybrid scaffold (HS) for AF tissue engineering. Methods The AYS was fabricated via conjugated electrospinning, while the aligned nanofibrous scaffold (AFS) was prepared by traditional electrospinning as a baseline scaffold. The HS was constructed by freeze-drying and cross-linking methods. Scanning electron microscopy and mechanical measurement were used to characterize the properties of these scaffolds. Bone marrow derived mesenchymal stem cells (BMSCs) were seeded on scaffolds, and cell proliferation was determined by CCK-8 assay, while cell infiltration and differentiation were assessed by histological measurement and quantitative real-time polymerase chain reaction, respectively. Results Morphological measurements showed that AYS presented a relatively better three-dimensional structure with larger pore sizes, higher porosity, and better fibers’ alignment compared to AFS. Mechanical testing demonstrated that the tensile property of AFS and AYS was qualitatively similar to the native AF tissue, albeit to a lesser extent. When BMSCs were seeded and cultured on these scaffolds, the number of cells cultured on HS and AYS was found to be significantly higher than that on AFS and culture plate after 7 days of culture (P<0.05). In addition, cell infiltration was significantly higher in HS when compared with AFS and AYS (P<0.05). A part of BMSCs ingressed into the inner part of AYS upon long-term in vitro culture. No significant difference was observed between AFS and AYS in terms of the median infiltration depth (P>0.05). BMSCs seeded on AYS demonstrated an increased expression of COL1A1, while the expression levels of SOX-9, COL2A1, and Aggrecan were higher in HS compared to other scaffolds (P<0.05). Conclusion These findings indicate that HS makes a proper scaffold for the AF tissue engineering as it replicates the axial compression and tensile property of AF, thereby providing a better platform for cell infiltration and cell–scaffold interaction.
Collapse
Affiliation(s)
- Jun Ma
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai
| | - Yunfei He
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai.,Department of Spinal Surgery, Lanzhou General Hospital of Lanzhou Military Command Region, Lanzhou
| | - Xilin Liu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai
| | - Weiming Chen
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University
| | - An Wang
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai.,Department of Orthopaedics, Shanghai Armed Police Force Hospital, Shanghai, China
| | - Chia-Ying Lin
- Structural Tissue Evaluation and Engineering Laboratories, Department of Biomedical Engineering.,Department of Orthopaedic Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Xiumei Mo
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University
| | - Xiaojian Ye
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai
| |
Collapse
|
35
|
Chu G, Shi C, Lin J, Wang S, Wang H, Liu T, Yang H, Li B. Biomechanics in Annulus Fibrosus Degeneration and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1078:409-420. [PMID: 30357635 DOI: 10.1007/978-981-13-0950-2_21] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Degenerative disc degeneration (DDD) is the major cause of low back pain, which seriously affects the life of patients. Current surgical and conservative treatments only relieve the pain temporarily, yet fail to restore the normal biomechanics and functions of healthy spine. Indeed, high recurrence of disc herniation commonly happens after discectomy. Degenerative changes in biomechanical and structural properties of the intervertebral disc (IVD), including fissures in annulus fibrosus (AF) and volume loss of nucleus pulposus (NP), mainly contribute to DDD development. AF plays a critical role in the biomechanical properties of IVD as it structural integrity is essential to confine NP and maintain physiological intradiscal pressure under loading. Maintaining the homeostasis of AF and NP, and thereby IVD, requires regulation of their biomechanics, which is also involved in the onset and subsequent development of AF degeneration. Therefore, it is essential to understand the biomechanical changes of AF during degeneration, which can also provide valuable insights into the repair and regeneration of AF. In this review, we focus on the biomechanical properties of AF tissue associated with its homeostasis and degeneration, and discuss the biomechanical stimulus required for regeneration of AF. We also provide an overview of recent strategies to target and modulate cell mechanics toward AF regeneration.
Collapse
Affiliation(s)
- Genglei Chu
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Chen Shi
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Jun Lin
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Shenghao Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Huan Wang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Tao Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Huilin Yang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Bin Li
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China. .,Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China. .,China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang, China.
| |
Collapse
|
36
|
Talò G, Turrisi C, Arrigoni C, Recordati C, Gerges I, Tamplenizza M, Cappelluti A, Riboldi S, Moretti M. Industrialization of a perfusion bioreactor: Prime example of a non‐straightforward process. J Tissue Eng Regen Med 2017; 12:405-415. [DOI: 10.1002/term.2480] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 05/05/2017] [Accepted: 05/09/2017] [Indexed: 01/17/2023]
Affiliation(s)
- G. Talò
- Cell and Tissue Engineering LaboratoryIRCCS Istituto Ortopedico Galeazzi Milan Italy
| | - C. Turrisi
- Dipartimento di Elettronica, Informazione e BioingegneriaPolitecnico di Milano Milan Italy
| | - C. Arrigoni
- Cell and Tissue Engineering LaboratoryIRCCS Istituto Ortopedico Galeazzi Milan Italy
| | | | | | | | - A. Cappelluti
- Fondazione Filarete Milan Italy
- SEMM European School of Molecular Medicine Milano Italy
| | | | - M. Moretti
- Cell and Tissue Engineering LaboratoryIRCCS Istituto Ortopedico Galeazzi Milan Italy
- Regenerative Medicine Technologies LaboratoryEnte Ospedaliero Cantonale (EOC) Lugano Switzerland
- Swiss Institute of Regenerative Medicine (SIRM) Torricella‐Taverne Switzerland
| |
Collapse
|
37
|
Vashaghian M, Zaat SJ, Smit TH, Roovers JP. Biomimetic implants for pelvic floor repair. Neurourol Urodyn 2017; 37:566-580. [PMID: 28799675 DOI: 10.1002/nau.23367] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/15/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Polypropylene implants are used for the reconstructive surgery of urogynaecological disorders like pelvic organ prolapse, but severe complications associated with their use have been reported. There is evidence that surface properties and a difference in mechanical stiffness between the implant and the host tissue contribute to these adverse events. Electrospinning is an innovative engineering alternative that provides a biomimetic microstructure for implants, resulting in a different mechano-biological performance. AIM The main objective of this review is to inform about the potential of electrospun matrices as an alternative modality for pelvic floor repair. METHODS Publications with the following studies of electrospun matrices were reviewed: (i) the technique; (ii) in vitro use for soft tissue engineering; (iii) in vivo use for reconstruction of soft tissues in animals; and (iv) clinical use in humans. RESULTS Electrospun matrices provide a synthetic mimic of natural extracellular matrix (ECM), favoring cellular attachment, proliferation and matrix deposition, through which a proper, low-inflammatory tissue-implant interaction can be established. Electrospun sheets can also be created with sufficient mechanical strength and stiffness for usage in prolapse surgery. CONCLUSION Electrospun matrices mimic the structural topography of the extracellular matrix and can be functionalized for better biological performance. As such, they have great potential for the next generation of urogynecological implants. However, their long-term safety and efficacy must still be established in vivo.
Collapse
Affiliation(s)
- Mahshid Vashaghian
- Department of Obstetrics & Gynaecology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sebastianus J Zaat
- Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Theodoor H Smit
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan-Paul Roovers
- Department of Obstetrics & Gynaecology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Kishan AP, Cosgriff-Hernandez EM. Recent advancements in electrospinning design for tissue engineering applications: A review. J Biomed Mater Res A 2017; 105:2892-2905. [DOI: 10.1002/jbm.a.36124] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/23/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Alysha P. Kishan
- Department of Biomedical Engineering; Texas A&M University, 5045 Emerging Technologies Building; 3120 TAMU College Station Texas 77843-3120
| | - Elizabeth M. Cosgriff-Hernandez
- Department of Biomedical Engineering; Texas A&M University, 5045 Emerging Technologies Building; 3120 TAMU College Station Texas 77843-3120
| |
Collapse
|
39
|
Font Tellado S, Bonani W, Balmayor ER, Foehr P, Motta A, Migliaresi C, van Griensven M. * Fabrication and Characterization of Biphasic Silk Fibroin Scaffolds for Tendon/Ligament-to-Bone Tissue Engineering. Tissue Eng Part A 2017; 23:859-872. [PMID: 28330431 DOI: 10.1089/ten.tea.2016.0460] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tissue engineering is an attractive strategy for tendon/ligament-to-bone interface repair. The structure and extracellular matrix composition of the interface are complex and allow for a gradual mechanical stress transfer between tendons/ligaments and bone. Thus, scaffolds mimicking the structural features of the native interface may be able to better support functional tissue regeneration. In this study, we fabricated biphasic silk fibroin scaffolds designed to mimic the gradient in collagen molecule alignment present at the interface. The scaffolds had two different pore alignments: anisotropic at the tendon/ligament side and isotropic at the bone side. Total porosity ranged from 50% to 80% and the majority of pores (80-90%) were <100-300 μm. Young's modulus varied from 689 to 1322 kPa depending on the type of construct. In addition, human adipose-derived mesenchymal stem cells were cultured on the scaffolds to evaluate the effect of pore morphology on cell proliferation and gene expression. Biphasic scaffolds supported cell attachment and influenced cytoskeleton organization depending on pore alignment. In addition, the gene expression of tendon/ligament, enthesis, and cartilage markers significantly changed depending on pore alignment in each region of the scaffolds. In conclusion, the biphasic scaffolds fabricated in this study show promising features for tendon/ligament-to-bone tissue engineering.
Collapse
Affiliation(s)
- Sònia Font Tellado
- 1 Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich , Munich, Germany
| | - Walter Bonani
- 2 Department of Industrial Engineering, BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Trento , Trento, Italy .,3 Trento Research Unit, INSTM-National Interuniversity Consortium of Materials Science and Technology , Trento, Italy
| | - Elizabeth R Balmayor
- 1 Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich , Munich, Germany
| | - Peter Foehr
- 4 Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich , Munich, Germany
| | - Antonella Motta
- 2 Department of Industrial Engineering, BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Trento , Trento, Italy
| | - Claudio Migliaresi
- 2 Department of Industrial Engineering, BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Trento , Trento, Italy .,3 Trento Research Unit, INSTM-National Interuniversity Consortium of Materials Science and Technology , Trento, Italy
| | - Martijn van Griensven
- 1 Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich , Munich, Germany
| |
Collapse
|
40
|
Tavakoli J. Tissue Engineering of the Intervertebral Disc's Annulus Fibrosus: A Scaffold-Based Review Study. Tissue Eng Regen Med 2017; 14:81-91. [PMID: 30603465 PMCID: PMC6171584 DOI: 10.1007/s13770-017-0024-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/10/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022] Open
Abstract
Tissue engineering as a high technology solution for treating disc's problem has been the focus of some researches recently; however, the upcoming successful results in this area depends on understanding the complexities of biology and engineering interface. Whereas the major responsibility of the nucleus pulposus is to provide a sustainable hydrated environment within the disc, the function of the annulus fibrosus (AF) is more mechanical, facilitating joint mobility and preventing radial bulging by confining of the central part, which makes the AF reconstruction important. Although the body of knowledge regarding the AF tissue engineering has grown rapidly, the opportunities to improve current understanding of how artificial scaffolds are able to mimic the AF concentric structure-including inter-lamellar matrix and cross-bridges-addressed unresolved research questions. The aim of this literature review was to collect and discuss, from the international scientific literature, information about tissue engineering of the AF based on scaffold fabrication and material properties, useful for developing new strategies in disc tissue engineering. The key parameter of this research was understanding if role of cross-bridges and inter-lamellar matrix has been considered on tissue engineering of the AF.
Collapse
Affiliation(s)
- Javad Tavakoli
- Medical Device Research Institute, School of Computer Science, Engineering and Mathematics, Flinders University, Adelaide, SA 5042 Australia
| |
Collapse
|
41
|
Pauly HM, Sathy BN, Olvera D, McCarthy HO, Kelly DJ, Popat KC, Dunne NJ, Haut Donahue TL. * Hierarchically Structured Electrospun Scaffolds with Chemically Conjugated Growth Factor for Ligament Tissue Engineering. Tissue Eng Part A 2017; 23:823-836. [PMID: 28350237 DOI: 10.1089/ten.tea.2016.0480] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The anterior cruciate ligament (ACL) of the knee is vital for proper joint function and is commonly ruptured during sports injuries or car accidents. Due to a lack of intrinsic healing capacity and drawbacks with allografts and autografts, there is a need for a tissue-engineered ACL replacement. Our group has previously used aligned sheets of electrospun polycaprolactone nanofibers to develop solid cylindrical bundles of longitudinally aligned nanofibers. We have shown that these nanofiber bundles support cell proliferation and elongation and the hierarchical structure and material properties are similar to the native human ACL. It is possible to combine multiple nanofiber bundles to create a scaffold that attempts to mimic the macroscale structure of the ACL. The goal of this work was to develop a hierarchical bioactive scaffold for ligament tissue engineering using connective tissue growth factor (CTGF)-conjugated nanofiber bundles and evaluate the behavior of mesenchymal stem cells (MSCs) on these scaffolds in vitro and in vivo. CTGF was immobilized onto the surface of individual nanofiber bundles or scaffolds consisting of multiple nanofiber bundles. The conjugation efficiency and the release of conjugated CTGF were assessed using X-ray photoelectron spectroscopy, assays, and immunofluorescence staining. Scaffolds were seeded with MSCs and maintained in vitro for 7 days (individual nanofiber bundles), in vitro for 21 days (scaled-up scaffolds of 20 nanofiber bundles), or in vivo for 6 weeks (small scaffolds of 4 nanofiber bundles), and ligament-specific tissue formation was assessed in comparison to non-CTGF-conjugated control scaffolds. Results showed that CTGF conjugation encouraged cell proliferation and ligament-specific tissue formation in vitro and in vivo. The results suggest that hierarchical electrospun nanofiber bundles conjugated with CTGF are a scalable and bioactive scaffold for ACL tissue engineering.
Collapse
Affiliation(s)
- Hannah M Pauly
- 1 School of Biomedical Engineering, Colorado State University , Fort Collins, Colorado
| | - Binulal N Sathy
- 2 Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute , Trinity College Dublin, Dublin, Ireland
| | - Dinorath Olvera
- 2 Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute , Trinity College Dublin, Dublin, Ireland
| | - Helen O McCarthy
- 3 School of Pharmacy, Queen's University Belfast , Belfast, United Kingdom
| | - Daniel J Kelly
- 2 Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute , Trinity College Dublin, Dublin, Ireland .,4 Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin , Dublin, Ireland .,5 Department of Anatomy, Royal College of Surgeons in Ireland , Dublin, Ireland .,6 Advanced Materials and Bioengineering Research Centre, Royal College of Surgeons in Ireland and Trinity College Dublin , Dublin, Ireland
| | - Ketul C Popat
- 1 School of Biomedical Engineering, Colorado State University , Fort Collins, Colorado.,7 Department of Mechanical Engineering, Colorado State University , Fort Collins, Colorado
| | - Nicholas J Dunne
- 2 Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute , Trinity College Dublin, Dublin, Ireland .,3 School of Pharmacy, Queen's University Belfast , Belfast, United Kingdom .,8 Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University , Dublin, Ireland
| | - Tammy Lynn Haut Donahue
- 1 School of Biomedical Engineering, Colorado State University , Fort Collins, Colorado.,7 Department of Mechanical Engineering, Colorado State University , Fort Collins, Colorado
| |
Collapse
|
42
|
O’Connell G, Garcia J, Amir J. 3D Bioprinting: New Directions in Articular Cartilage Tissue Engineering. ACS Biomater Sci Eng 2017; 3:2657-2668. [DOI: 10.1021/acsbiomaterials.6b00587] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Grace O’Connell
- Department
of Mechanical Engineering University of California, Berkeley, 5122 Etcheverry Hall, Berkeley, California 94720, United States
| | - Jeanette Garcia
- IBM Research-Almaden, 650
Harry Road K17/D2, San Jose, California 95120, United States
| | - Jamali Amir
- Joint Preservation Institute, 2825 J Street #440, Sacramento, California 95816, United States
| |
Collapse
|
43
|
Long RG, Torre OM, Hom WW, Assael DJ, Iatridis JC. Design Requirements for Annulus Fibrosus Repair: Review of Forces, Displacements, and Material Properties of the Intervertebral Disk and a Summary of Candidate Hydrogels for Repair. J Biomech Eng 2016; 138:021007. [PMID: 26720265 DOI: 10.1115/1.4032353] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Indexed: 02/02/2023]
Abstract
There is currently a lack of clinically available solutions to restore functionality to the intervertebral disk (IVD) following herniation injury to the annulus fibrosus (AF). Microdiscectomy is a commonly performed surgical procedure to alleviate pain caused by herniation; however, AF defects remain and can lead to accelerated degeneration and painful conditions. Currently available AF closure techniques do not restore mechanical functionality or promote tissue regeneration, and have risk of reherniation. This review determined quantitative design requirements for AF repair materials and summarized currently available hydrogels capable of meeting these design requirements by using a series of systematic PubMed database searches to yield 1500+ papers that were screened and analyzed for relevance to human lumbar in vivo measurements, motion segment behaviors, and tissue level properties. We propose a testing paradigm involving screening tests as well as more involved in situ and in vivo validation tests to efficiently identify promising biomaterials for AF repair. We suggest that successful materials must have high adhesion strength (∼0.2 MPa), match as many AF material properties as possible (e.g., approximately 1 MPa, 0. 3 MPa, and 30 MPa for compressive, shear, and tensile moduli, respectively), and have high tensile failure strain (∼65%) to advance to in situ and in vivo validation tests. While many biomaterials exist for AF repair, few undergo extensive mechanical characterization. A few hydrogels show promise for AF repair since they can match at least one material property of the AF while also adhering to AF tissue and are capable of easy implantation during surgical procedures to warrant additional optimization and validation.
Collapse
|
44
|
Biazar E. Application of polymeric nanofibers in medical designs, part III: Musculoskeletal and urological tissues. INT J POLYM MATER PO 2016. [DOI: 10.1080/00914037.2016.1180620] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Esmaeil Biazar
- Department of Biomaterials Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| |
Collapse
|
45
|
D'Amore A, Soares JS, Stella JA, Zhang W, Amoroso NJ, Mayer JE, Wagner WR, Sacks MS. Large strain stimulation promotes extracellular matrix production and stiffness in an elastomeric scaffold model. J Mech Behav Biomed Mater 2016; 62:619-635. [PMID: 27344402 PMCID: PMC4955736 DOI: 10.1016/j.jmbbm.2016.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/30/2016] [Accepted: 05/03/2016] [Indexed: 01/07/2023]
Abstract
Mechanical conditioning of engineered tissue constructs is widely recognized as one of the most relevant methods to enhance tissue accretion and microstructure, leading to improved mechanical behaviors. The understanding of the underlying mechanisms remains rather limited, restricting the development of in silico models of these phenomena, and the translation of engineered tissues into clinical application. In the present study, we examined the role of large strip-biaxial strains (up to 50%) on ECM synthesis by vascular smooth muscle cells (VSMCs) micro-integrated into electrospun polyester urethane urea (PEUU) constructs over the course of 3 weeks. Experimental results indicated that VSMC biosynthetic behavior was quite sensitive to tissue strain maximum level, and that collagen was the primary ECM component synthesized. Moreover, we found that while a 30% peak strain level achieved maximum ECM synthesis rate, further increases in strain level lead to a reduction in ECM biosynthesis. Subsequent mechanical analysis of the formed collagen fiber network was performed by removing the scaffold mechanical responses using a strain-energy based approach, showing that the denovo collagen also demonstrated mechanical behaviors substantially better than previously obtained with small strain training and comparable to mature collagenous tissues. We conclude that the application of large deformations can play a critical role not only in the quantity of ECM synthesis (i.e. the rate of mass production), but also on the modulation of the stiffness of the newly formed ECM constituents. The improved understanding of the process of growth and development of ECM in these mechano-sensitive cell-scaffold systems will lead to more rational design and manufacturing of engineered tissues operating under highly demanding mechanical environments.
Collapse
Affiliation(s)
- Antonio D'Amore
- Department of Bioengineering McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Fondazione RiMED, Italy; DICGIM, Università di Palermo, Italy
| | - Joao S Soares
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences, Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - John A Stella
- Department of Bioengineering McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Will Zhang
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences, Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas J Amoroso
- Department of Bioengineering McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John E Mayer
- Department of Cardiac Surgery Boston Children׳s Hospital and Harvard Medical School, Boston, MA, USA
| | - William R Wagner
- Department of Bioengineering McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael S Sacks
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences, Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
46
|
Mechanical properties and cellular response of novel electrospun nanofibers for ligament tissue engineering: Effects of orientation and geometry. J Mech Behav Biomed Mater 2016; 61:258-270. [DOI: 10.1016/j.jmbbm.2016.03.022] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 03/13/2016] [Accepted: 03/21/2016] [Indexed: 01/13/2023]
|
47
|
Advances in combining gene therapy with cell and tissue engineering-based approaches to enhance healing of the meniscus. Osteoarthritis Cartilage 2016; 24:1330-9. [PMID: 27063441 PMCID: PMC5298218 DOI: 10.1016/j.joca.2016.03.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/17/2016] [Accepted: 03/25/2016] [Indexed: 02/02/2023]
Abstract
Meniscal lesions are common problems in orthopaedic surgery and sports medicine, and injury or loss of the meniscus accelerates the onset of knee osteoarthritis (OA). Despite a variety of therapeutic options in the clinics, there is a critical need for improved treatments to enhance meniscal repair. In this regard, combining gene-, cell-, and tissue engineering-based approaches is an attractive strategy to generate novel, effective therapies to treat meniscal lesions. In the present work, we provide an overview of the tools currently available to improve meniscal repair and discuss the progress and remaining challenges for potential future translation in patients.
Collapse
|
48
|
Wu J, Hong Y. Enhancing cell infiltration of electrospun fibrous scaffolds in tissue regeneration. Bioact Mater 2016; 1:56-64. [PMID: 29744395 PMCID: PMC5883964 DOI: 10.1016/j.bioactmat.2016.07.001] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/02/2016] [Accepted: 07/13/2016] [Indexed: 12/30/2022] Open
Abstract
Electrospinning is one of the most effective approaches to fabricate tissue-engineered scaffolds composed of nano-to sub-microscale fibers that simulate a native extracellular matrix. However, one major concern about electrospun scaffolds for tissue repair and regeneration is that their small pores defined by densely compacted fibers markedly hinder cell infiltration and tissue ingrowth. To address this problem, researchers have developed and investigated various methods of manipulating scaffold structures to increase pore size or loosen the scaffold. These methods involve the use of physical treatments, such as salt leaching, gas foaming and custom-made collectors, and combined techniques to obtain electrospun scaffolds with loose fibrous structures and large pores. This article provides a summary of these motivating electrospinning techniques to enhance cell infiltration of electrospun scaffolds, which may inspire new electrospinning techniques and their new biomedical applications. Electrospinning is a popular and attractive technique to produce fibrous scaffolds for tissue regeneration. One limitation for electrospun scaffolds is low cell infiltration. This article summarizes innovative techniques to improve cell infiltration of electrospun scaffolds.
Collapse
Affiliation(s)
- Jinglei Wu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA.,Joint Graduate Program in Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA.,Joint Graduate Program in Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
49
|
Cell-Based Strategies for Meniscus Tissue Engineering. Stem Cells Int 2016; 2016:4717184. [PMID: 27274735 PMCID: PMC4871968 DOI: 10.1155/2016/4717184] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/06/2016] [Accepted: 02/11/2016] [Indexed: 12/14/2022] Open
Abstract
Meniscus injuries remain a significant challenge due to the poor healing potential of the inner avascular zone. Following a series of studies and clinical trials, tissue engineering is considered a promising prospect for meniscus repair and regeneration. As one of the key factors in tissue engineering, cells are believed to be highly beneficial in generating bionic meniscus structures to replace injured ones in patients. Therefore, cell-based strategies for meniscus tissue engineering play a fundamental role in meniscal regeneration. According to current studies, the main cell-based strategies for meniscus tissue engineering are single cell type strategies; cell coculture strategies also were applied to meniscus tissue engineering. Likewise, on the one side, the zonal recapitulation strategies based on mimicking meniscal differing cells and internal architectures have received wide attentions. On the other side, cell self-assembling strategies without any scaffolds may be a better way to build a bionic meniscus. In this review, we primarily discuss cell seeds for meniscus tissue engineering and their application strategies. We also discuss recent advances and achievements in meniscus repair experiments that further improve our understanding of meniscus tissue engineering.
Collapse
|
50
|
Fabrication and detection of tissue engineered bone aggregates based on encapsulated human ADSCs within hybrid calcium alginate/bone powder gel-beads in a spinner flask. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 62:787-94. [DOI: 10.1016/j.msec.2016.02.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/27/2016] [Accepted: 02/11/2016] [Indexed: 12/31/2022]
|