1
|
Xu H, Tang Y, Mao W, Wu L, Zhou Y, Deng J, Tang W, Xiao X, Xia Y, Wang Y. Exploration of the optimal retention method in vivo for stem cell therapy: Low-intensity ultrasound preconditioning. Regen Ther 2025; 29:484-492. [PMID: 40390863 PMCID: PMC12088760 DOI: 10.1016/j.reth.2025.04.012] [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: 01/10/2025] [Revised: 03/26/2025] [Accepted: 04/17/2025] [Indexed: 05/21/2025] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are pluripotent and self-renewing, exerting a crucial role in the domain of regenerative medicine. Nevertheless, BMSCs encounter challenges such as low cell viability and inadequate homing during transplantation, thereby restricting their therapeutic efficacy. Hence, current research is concentrated on identifying optimal retention approaches following BMSCs transplantation to enhance its effectiveness. Low-intensity ultrasound (LIUS) has been verified as an efficacious method to enhance the performance of BMSCs. We established a skin trauma model and assessed the therapeutic effect of LIUS-preconditioned BMSCs. The results demonstrated that pretreatment with LIUS could expedite wound healing and effectively diminish scar formation post-transplantation by promoting proliferation capacity, reinforcing anti-apoptotic attributes, improving homing ability, and significantly enhancing the transplantation effect of BMSCs. These discoveries imply that LIUS might constitute a promising strategy for attaining optimal retention after stem cell transplantation in regenerative medicine and wound repair therapy.
Collapse
Affiliation(s)
| | | | - Wenjing Mao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Liu Wu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yiqing Zhou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Juan Deng
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Wentao Tang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Xinfang Xiao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yi Xia
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yan Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
2
|
Song W, Zhao D, Wang J, Han Z, Liu Y, Wang Y, Yang C. Ultrasound-Driven Innervated Bone Regeneration in Additively Manufactured Degradable Metallic Scaffolds. Adv Healthc Mater 2025; 14:e2404024. [PMID: 40152173 DOI: 10.1002/adhm.202404024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Bone tissues are densely innervated by nerve fibers throughout the periosteum and mineralized bone. The impairment of innervated bone regeneration is a critical factor contributing to the challenges in osteoporotic bone remodeling and repair. Herein, an "ultrasound-driven innervated bone regeneration" strategy is proposed in additively manufactured degradable Zn-Cu scaffolds. The in vitro investigations with RSC96 cells elucidated the synergistic promotion of low-intensity pulsed ultrasound (LIPUS) and metal cations on Schwann cell proliferation and exosome secretion. Notably, these Schwann cell-derived exosomes, once internalized by neighboring bone marrow stromal cells (BMSCs), significantly enhanced their migration, osteogenic differentiation, and extracellular matrix deposition, indicating a potent mechanism for innervated bone regeneration. Furthermore, the in vivo evaluation validated that LIPUS stimulation significantly activated S100β-positive Schwann cells and facilitated the regeneration of peripheral nerve fibers within cranial defects, leading to accelerated bone healing of osteoporotic rats with Zn-Cu implantation over 2- and 6-week recovery periods. This work provides an innervated bone regeneration strategy by focusing on the activation of Schwann cells and enhancement of paracrine effect, especially exosome secretion, which further recruited surrounding BMSCs and promoted their osteogenic differentiation. This study holds considerable promise for clinical applications and translation in the treatment of osteoporotic bone defects.
Collapse
Affiliation(s)
- Wencheng Song
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Danlei Zhao
- State Key Laboratory of High-performance Precision Manufacturing, Dalian University of Technology, Dalian, 116023, China
| | - Jiajia Wang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Zhengshuo Han
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yijun Liu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yifan Wang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Cheng Yang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| |
Collapse
|
3
|
Li Z, Su T, Yang Y, Zhao H. Construction of Multicellular Neural Tissue Using Three-Dimensional Printing Technology: Cell Interaction. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40256794 DOI: 10.1089/ten.teb.2024.0323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
The study of the human nervous system remains challenging due to its inherent complexity and difficulty in obtaining original samples. Three-dimensional (3D) bioprinting is a rapidly evolving technology in the field of tissue engineering that has made significant contributions to several disciplines, including neuroscience. In order to more accurately reflect the intricate multicellular milieu of the in vivo environment, an increasing number of studies have commenced experimentation with the coprinting of diverse cell types. This article provides an overview of technical details and the application of 3D bioprinting with multiple cell types in the field of neuroscience, focusing on the challenges of coprinting and the research conducted based on multicellular printing. This review discusses cell interactions in coprinting systems, stem cell applications, the construction of brain-like organoids, the establishment of disease models, and the potential for integrating 3D bioprinting with other 3D culture techniques.
Collapse
Affiliation(s)
- Zhixiang Li
- Tissue Engineering Laboratory, School of Biology, Food, and Environment, Hefei University, Hefei, PR China
| | - Tong Su
- Tissue Engineering Laboratory, School of Biology, Food, and Environment, Hefei University, Hefei, PR China
| | - Yujie Yang
- Tissue Engineering Laboratory, School of Biology, Food, and Environment, Hefei University, Hefei, PR China
| | - Huan Zhao
- Tissue Engineering Laboratory, School of Biology, Food, and Environment, Hefei University, Hefei, PR China
| |
Collapse
|
4
|
Liu X, Feng Z, Ran Z, Zeng Y, Cao G, Li X, Ye H, Wang M, Liang W, He Y. External Stimuli-Responsive Strategies for Surface Modification of Orthopedic Implants: Killing Bacteria and Enhancing Osteogenesis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67028-67044. [PMID: 38497341 DOI: 10.1021/acsami.3c19149] [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: 03/19/2024]
Abstract
Bacterial infection and insufficient osteogenic activity are the main causes of orthopedic implant failure. Conventional surface modification methods are difficult to meet the requirements for long-term implant placement. In order to better regulate the function of implant surfaces, especially to improve both the antibacterial and osteogenic activity, external stimuli-responsive (ESR) strategies have been employed for the surface modification of orthopedic implants. External stimuli act as "smart switches" to regulate the surface interactions with bacteria and cells. The balance between antibacterial and osteogenic capabilities of implant surfaces can be achieved through these specific ESR manifestations, including temperature changes, reactive oxygen species production, controlled release of bioactive molecules, controlled release of functional ions, etc. This Review summarizes the recent progress on different ESR strategies (based on light, ultrasound, electric, and magnetic fields) that can effectively balance antibacterial performance and osteogenic capability of orthopedic implants. Furthermore, the current limitations and challenges of ESR strategies for surface modification of orthopedic implants as well as future development direction are also discussed.
Collapse
Affiliation(s)
- Xujie Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhenzhen Feng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhili Ran
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yaoxun Zeng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Guining Cao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xinyi Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Huiling Ye
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Meijing Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Wanting Liang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yan He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
5
|
Marcotulli M, Barbetta A, Scarpa E, Bini F, Marinozzi F, Ruocco G, Casciola CM, Scognamiglio C, Carugo D, Cidonio G. Jingle Cell Rock: Steering Cellular Activity With Low-Intensity Pulsed Ultrasound (LIPUS) to Engineer Functional Tissues in Regenerative Medicine. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1973-1986. [PMID: 39289118 DOI: 10.1016/j.ultrasmedbio.2024.08.016] [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: 03/15/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024]
Abstract
Acoustic manipulation or perturbation of biological soft matter has emerged as a promising clinical treatment for a number of applications within regenerative medicine, ranging from bone fracture repair to neuromodulation. The potential of ultrasound (US) endures in imparting mechanical stimuli that are able to trigger a cascade of molecular signals within unscathed cells. Particularly, low-intensity pulsed ultrasound (LIPUS) has been associated with bio-effects such as activation of specific cellular pathways and alteration of cell morphology and gene expression, the extent of which can be modulated by fine tuning of LIPUS parameters including intensity, frequency and exposure time. Although the molecular mechanisms underlying LIPUS are not yet fully elucidated, a number of studies clearly define the modulation of specific ultrasonic parameters as a means to guide the differentiation of a specific set of stem cells towards adult and fully differentiated cell types. Herein, we outline the applications of LIPUS in regenerative medicine and the in vivo and in vitro studies that have confirmed the unbounded clinical potential of this platform. We highlight the latest developments aimed at investigating the physical and biological mechanisms of action of LIPUS, outlining the most recent efforts in using this technology to aid tissue engineering strategies for repairing tissue or modelling specific diseases. Ultimately, we detail tissue-specific applications harnessing LIPUS stimuli, offering insights over the engineering of new constructs and therapeutic modalities. Overall, we aim to lay the foundation for a deeper understanding of the mechanisms governing LIPUS-based therapy, to inform the development of safer and more effective tissue regeneration strategies in the field of regenerative medicine.
Collapse
Affiliation(s)
- Martina Marcotulli
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy; Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy; Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Andrea Barbetta
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Edoardo Scarpa
- Infection Dynamics Laboratory, Department of Pharmaceutical Sciences, University of Milan, Milan, Italy; National Institute of Molecular Gentics (INGM), Milan, Italy; Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Fabiano Bini
- Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy
| | - Franco Marinozzi
- Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ruocco
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy
| | - Carlo Massimo Casciola
- Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy
| | - Chiara Scognamiglio
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy
| | - Dario Carugo
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Gianluca Cidonio
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy; Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK; Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
6
|
Melo WGGD, Bezerra DDO, Silva ERDDFS, Campêlo CB, Carvalho MAMD, Argôlo Neto NM. Behavioral dynamics of medicinal signaling cells from porcine bone marrow in long-term culture. Can J Physiol Pharmacol 2024; 102:672-679. [PMID: 39189463 DOI: 10.1139/cjpp-2023-0458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Medicinal signaling cells (MSC) hold promise for regenerative medicine due to their ability to repair damaged tissues. However, their effectiveness can be affected by how long they are cultured in the lab. This study investigated how passage number influences key properties for regenerative medicine of pig bone marrow MSC. The medicinal signiling cells derived from pig bone marrow (BM-MSC) were cultured in D-MEM High Glucose supplemented with 15% foetal bovine serum until the 25th passage and assessed their growth, viability, ability to differentiate into different cell types (plasticity), and cell cycle activity. Our findings showed that while the cells remained viable until the 25th passage, their ability to grow and differentiate declined after the 5th passage. Additionally, cells in later passages spent more time in a resting phase, suggesting reduced activity. In conclusion, the number of passages is a critical factor for maintaining ideal MSC characteristics. From the 9th passage BM-MSC exhibit decline in proliferation, differentiation potential, and cell cycle activity. Given this, it is possible to suggest that the use of 5th passage cells is the most suitable for therapeutic applications.
Collapse
Affiliation(s)
- Wanderson Gabriel Gomes de Melo
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Dayseanny de Oliveira Bezerra
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | | | - Camile Benício Campêlo
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Maria Acelina Martins de Carvalho
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Napoleão Martins Argôlo Neto
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| |
Collapse
|
7
|
Zhou J, Ning E, Lu L, Zhang H, Yang X, Hao Y. Effectiveness of low-intensity pulsed ultrasound on osteoarthritis: molecular mechanism and tissue engineering. Front Med (Lausanne) 2024; 11:1292473. [PMID: 38695024 PMCID: PMC11061361 DOI: 10.3389/fmed.2024.1292473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/05/2024] [Indexed: 05/04/2024] Open
Abstract
Osteoarthritis (OA) is distinguished by pathological alterations in the synovial membrane, articular cartilage, and subchondral bone, resulting in physical symptoms such as pain, deformity, and impaired mobility. Numerous research studies have validated the effectiveness of low-intensity pulsed ultrasound (LIPUS) in OA treatment. The periodic mechanical waves generated by LIPUS can mitigate cellular ischemia and hypoxia, induce vibration and collision, produce notable thermal and non-thermal effects, alter cellular metabolism, expedite tissue repair, improve nutrient delivery, and accelerate the healing process of damaged tissues. The efficacy and specific mechanism of LIPUS is currently under investigation. This review provides an overview of LIPUS's potential role in the treatment of OA, considering various perspectives such as the synovial membrane, cartilage, subchondral bone, and tissue engineering. It aims to facilitate interdisciplinary scientific research and further exploration of LIPUS as a complementary technique to existing methods or surgery. Ongoing research is focused on determining the optimal dosage, frequency, timing, and treatment strategy of LIPUS for OA. Additional research is required to clarify the precise mechanism of action and potential impacts on cellular, animal, and human systems prior to its integration into therapeutic applications.
Collapse
Affiliation(s)
- Jing Zhou
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Eryu Ning
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Lingfeng Lu
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Huili Zhang
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
8
|
Wu T, Zheng F, Tang HY, Li HZ, Cui XY, Ding S, Liu D, Li CY, Jiang JH, Yang RL. Low-intensity pulsed ultrasound reduces alveolar bone resorption during orthodontic treatment via Lamin A/C-Yes-associated protein axis in stem cells. World J Stem Cells 2024; 16:267-286. [PMID: 38577236 PMCID: PMC10989285 DOI: 10.4252/wjsc.v16.i3.267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/30/2023] [Accepted: 02/01/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The bone remodeling during orthodontic treatment for malocclusion often requires a long duration of around two to three years, which also may lead to some complications such as alveolar bone resorption or tooth root resorption. Low-intensity pulsed ultrasound (LIPUS), a noninvasive physical therapy, has been shown to promote bone fracture healing. It is also reported that LIPUS could reduce the duration of orthodontic treatment; however, how LIPUS regulates the bone metabolism during the orthodontic treatment process is still unclear. AIM To investigate the effects of LIPUS on bone remodeling in an orthodontic tooth movement (OTM) model and explore the underlying mechanisms. METHODS A rat model of OTM was established, and alveolar bone remodeling and tooth movement rate were evaluated via micro-computed tomography and staining of tissue sections. In vitro, human bone marrow mesenchymal stem cells (hBMSCs) were isolated to detect their osteogenic differentiation potential under compression and LIPUS stimulation by quantitative reverse transcription-polymerase chain reaction, Western blot, alkaline phosphatase (ALP) staining, and Alizarin red staining. The expression of Yes-associated protein (YAP1), the actin cytoskeleton, and the Lamin A/C nucleoskeleton were detected with or without YAP1 small interfering RNA (siRNA) application via immunofluorescence. RESULTS The force treatment inhibited the osteogenic differentiation potential of hBMSCs; moreover, the expression of osteogenesis markers, such as type 1 collagen (COL1), runt-related transcription factor 2, ALP, and osteocalcin (OCN), decreased. LIPUS could rescue the osteogenic differentiation of hBMSCs with increased expression of osteogenic marker inhibited by force. Mechanically, the expression of LaminA/C, F-actin, and YAP1 was downregulated after force treatment, which could be rescued by LIPUS. Moreover, the osteogenic differentiation of hBMSCs increased by LIPUS could be attenuated by YAP siRNA treatment. Consistently, LIPUS increased alveolar bone density and decreased vertical bone absorption in vivo. The decreased expression of COL1, OCN, and YAP1 on the compression side of the alveolar bone was partially rescued by LIPUS. CONCLUSION LIPUS can accelerate tooth movement and reduce alveolar bone resorption by modulating the cytoskeleton-Lamin A/C-YAP axis, which may be a promising strategy to reduce the orthodontic treatment process.
Collapse
Affiliation(s)
- Tong Wu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Fu Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Hong-Yi Tang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Hua-Zhi Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Xin-Yu Cui
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Shuai Ding
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Duo Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Cui-Ying Li
- Department of Central Laboratory, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Jiu-Hui Jiang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Rui-Li Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China.
| |
Collapse
|
9
|
He YF, Wang XL, Deng SP, Wang YL, Huang QQ, Lin S, Lyu GR. Latest progress in low-intensity pulsed ultrasound for studying exosomes derived from stem/progenitor cells. Front Endocrinol (Lausanne) 2023; 14:1286900. [PMID: 38089611 PMCID: PMC10715436 DOI: 10.3389/fendo.2023.1286900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Stem cells have self-renewal, replication, and multidirectional differentiation potential, while progenitor cells are undifferentiated, pluripotent or specialized stem cells. Stem/progenitor cells secrete various factors, such as cytokines, exosomes, non-coding RNAs, and proteins, and have a wide range of applications in regenerative medicine. However, therapies based on stem cells and their secreted exosomes present limitations, such as insufficient source materials, mature differentiation, and low transplantation success rates, and methods addressing these problems are urgently required. Ultrasound is gaining increasing attention as an emerging technology. Low-intensity pulsed ultrasound (LIPUS) has mechanical, thermal, and cavitation effects and produces vibrational stimuli that can lead to a series of biochemical changes in organs, tissues, and cells, such as the release of extracellular bodies, cytokines, and other signals. These changes can alter the cellular microenvironment and affect biological behaviors, such as cell differentiation and proliferation. Here, we discuss the effects of LIPUS on the biological functions of stem/progenitor cells, exosomes, and non-coding RNAs, alterations involved in related pathways, various emerging applications, and future perspectives. We review the roles and mechanisms of LIPUS in stem/progenitor cells and exosomes with the aim of providing a deeper understanding of LIPUS and promoting research and development in this field.
Collapse
Affiliation(s)
- Yi-fang He
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xia-li Wang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Departments of Medical Imaging, Quanzhou Medical College, Quanzhou, China
| | - Shuang-ping Deng
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan-li Wang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qing-qing Huang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia
| | - Guo-rong Lyu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Departments of Medical Imaging, Quanzhou Medical College, Quanzhou, China
| |
Collapse
|
10
|
Huang Y, Zhang Z, Bi F, Tang H, Chen J, Huo F, Chen J, Lan T, Qiao X, Sima X, Guo W. Personalized 3D-Printed Scaffolds with Multiple Bioactivities for Bioroot Regeneration. Adv Healthc Mater 2023; 12:e2300625. [PMID: 37523260 DOI: 10.1002/adhm.202300625] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/26/2023] [Indexed: 08/02/2023]
Abstract
Recent advances in 3D printing offer a prospective avenue for producing transplantable human tissues with complex geometries; however, the appropriate 3D-printed scaffolds possessing the biological compatibility for tooth regeneration remain unidentified. This study proposes a personalized scaffold of multiple bioactivities, including induction of stem cell proliferation and differentiation, biomimetic mineralization, and angiogenesis. A brand-new bioink system comprising a biocompatible and biodegradable polymer is developed and reinforced with extracellular matrix generated from dentin tissue (treated dentin matrix, TDM). Adding TDM optimizes physical properties including microstructure, hydrophilicity, and mechanical strength of the scaffolds. Proteomics analysis reveals that the released proteins of the 3D-printed TDM scaffolds relate to multiple biological processes and interact closely with each other. Additionally, 3D-printed TDM scaffolds establish a favorable microenvironment for cell attachment, proliferation, and differentiation in vitro. The 3D-printed TDM scaffolds are proangiogenic and facilitate whole-thickness vascularization of the graft in a subcutaneous model. Notably, the personalized TDM scaffold combined with dental follicle cells mimics the anatomy and physiology of the native tooth root three months after in situ transplantation in beagles. The remarkable in vitro and in vivo outcomes suggest that the 3D-printed TDM scaffolds have multiple bioactivities and immense clinical potential for tooth-loss therapy.
Collapse
Affiliation(s)
- Yibing Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhijun Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Fei Bi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Huilin Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Jiahao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Fangjun Huo
- State Key Laboratory of Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Jie Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Tingting Lan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Xiangchen Qiao
- Chengdu Guardental Technology Limited Corporation, Chengdu, 610041, P. R. China
| | - Xiutian Sima
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Weihua Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
- Yunnan Key Laboratory of Stomatology, Affiliated Hospital of Stomatology, School of Stomatology, Kunming Medical University, Kunming, 650000, P. R. China
| |
Collapse
|
11
|
Aliabouzar M, Quesada C, Chan ZQ, Fowlkes JB, Franceschi RT, Putnam AJ, Fabiilli ML. Acoustic droplet vaporization for on-demand modulation of microporosity in smart hydrogels. Acta Biomater 2023; 164:195-208. [PMID: 37121372 PMCID: PMC10538466 DOI: 10.1016/j.actbio.2023.04.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/10/2023] [Accepted: 04/25/2023] [Indexed: 05/02/2023]
Abstract
Microporosity in hydrogels is critical for directing tissue formation and function. We have developed a fibrin-based smart hydrogel, termed an acoustically responsive scaffold (ARS), which responds to focused ultrasound in a spatiotemporally controlled, user-defined manner. ARSs are highly flexible platforms due to the inclusion of phase-shift droplets and their tunable response to ultrasound through a mechanism termed acoustic droplet vaporization (ADV). Here, we demonstrated that ADV enabled consistent generation of micropores in ARSs, throughout the entire thickness (∼5.5 mm), utilizing perfluorooctane phase-shift droplets. Size characteristics of the generated micropores were quantified in response to critical parameters including acoustic properties, droplet size, and shear elastic modulus of fibrin using confocal microscopy. The findings showed that the length of the generated micropores correlated directly with excitation frequency, peak rarefactional pressure, pulse duration, droplet size, and indirectly with the shear elastic modulus of the fibrin matrix. The ADV-generated micropores in ARSs were further compared with cavitation-mediated micropores in fibrin gels without droplets. Additionally, the Keller-Miksis equation was used to predict an upper bound for micropore formation in ARSs at varying driving frequencies and droplet sizes. Finally, our in vivo studies showed that host cell migration following ADV-induced micropore formation was frequency-dependent, with up to 2.6 times higher cell migration at lower frequencies. Overall, these findings demonstrate a new potential application of ADV in hydrogels. STATEMENT OF SIGNIFICANCE: Interconnected micropores within a hydrogel can facilitate many cell-mediated processes. Most techniques for generating micropores are typically not biocompatible or do not enable controlled, in situ micropore formation. We used an ultrasound-based technique, termed acoustic droplet vaporization, to generate microporosity in smart hydrogels termed acoustically responsive scaffolds (ARSs). ARSs contain a fibrin matrix doped with a phase-shift droplet. We demonstrate that unique acoustic properties of phase-shift droplets can be tailored to yield spatiotemporally controlled, on-demand micropore formation. Additionally, the size characteristics of the ultrasound-generated micropores can be modulated by tuning ultrasound parameters, droplet properties, and bulk elastic properties of fibrin. Finally, we demonstrate significant, frequency-dependent host cell migration in subcutaneously implanted ARSs in mice following ultrasound-induced micropore formation in situ.
Collapse
Affiliation(s)
- Mitra Aliabouzar
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | - Carole Quesada
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Ze Qi Chan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - J Brian Fowlkes
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| | - Renny T Franceschi
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Dental School, University of Michigan, Ann Arbor, MI, USA
| | - Andrew J Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mario L Fabiilli
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
12
|
Jin L, Shan J, Hao Y, Wang Y, Liu L. Enhanced bone regeneration by low-intensity pulsed ultrasound and lipid microbubbles on PLGA/TCP 3D-printed scaffolds. BMC Biotechnol 2023; 23:13. [PMID: 37280578 DOI: 10.1186/s12896-023-00783-9] [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: 12/03/2022] [Accepted: 05/23/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND To investigate the effect of low-intensity pulsed ultrasound (LIPUS) combined with lipid microbubbles on the proliferation and bone regeneration of bone marrow mesenchymal stem cells (BMSCs) in poly (lactic-glycolic acid copolymer) (PLGA)/α-tricalcium phosphate (TCP) 3D-printed scaffolds. METHODS BMSCs were irradiated with different LIPUS parameters and microbubble concentrations, and the best acoustic excitation parameters were selected. The expression of type I collagen and the activity of alkaline phosphatase were detected. Alizarin red staining was used to evaluate the calcium salt production during osteogenic differentiation. RESULTS BMSCs proliferation was the most significant under the condition of 0.5% (v/v) lipid microbubble concentration, 2.0 MHz frequency, 0.3 W/cm2 sound intensity and 20% duty cycle. After 14 days, the type I collagen expression and alkaline phosphatase activity in the scaffold increased significantly compared to those in the control group, and alizarin red staining showed more calcium salt production during osteogenic differentiation. After 21 days, scanning electron microscopy experiments showed that osteogenesis was obvious in the PLGA/TCP scaffolds. CONCLUSION LIPUS combined with lipid microbubbles on PLGA/TCP scaffolds can promote BMSCs growth and bone differentiation, which is expected to provide a new and effective method for the treatment of bone regeneration in tissue engineering.
Collapse
Affiliation(s)
- Lin Jin
- Department of Ultrasound, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
- Department of Ultrasound, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Jiali Shan
- Department of Ultrasound, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - Yanhong Hao
- Department of Ultrasound, First Hospital of Shanxi Medical University, No. 85, Jiefang South Road, Taiyuan, 030001, Shanxi, China
| | - Yingchun Wang
- Department of Ultrasound, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - Liping Liu
- Department of Ultrasound, First Hospital of Shanxi Medical University, No. 85, Jiefang South Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
13
|
Ambattu LA, Yeo LY. Sonomechanobiology: Vibrational stimulation of cells and its therapeutic implications. BIOPHYSICS REVIEWS 2023; 4:021301. [PMID: 38504927 PMCID: PMC10903386 DOI: 10.1063/5.0127122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/27/2023] [Indexed: 03/21/2024]
Abstract
All cells possess an innate ability to respond to a range of mechanical stimuli through their complex internal machinery. This comprises various mechanosensory elements that detect these mechanical cues and diverse cytoskeletal structures that transmit the force to different parts of the cell, where they are transcribed into complex transcriptomic and signaling events that determine their response and fate. In contrast to static (or steady) mechanostimuli primarily involving constant-force loading such as compression, tension, and shear (or forces applied at very low oscillatory frequencies (≤ 1 Hz) that essentially render their effects quasi-static), dynamic mechanostimuli comprising more complex vibrational forms (e.g., time-dependent, i.e., periodic, forcing) at higher frequencies are less well understood in comparison. We review the mechanotransductive processes associated with such acoustic forcing, typically at ultrasonic frequencies (> 20 kHz), and discuss the various applications that arise from the cellular responses that are generated, particularly for regenerative therapeutics, such as exosome biogenesis, stem cell differentiation, and endothelial barrier modulation. Finally, we offer perspectives on the possible existence of a universal mechanism that is common across all forms of acoustically driven mechanostimuli that underscores the central role of the cell membrane as the key effector, and calcium as the dominant second messenger, in the mechanotransduction process.
Collapse
Affiliation(s)
- Lizebona August Ambattu
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne VIC 3000, Australia
| | - Leslie Y. Yeo
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne VIC 3000, Australia
| |
Collapse
|
14
|
Chen Y, Yang H, Wang Z, Zhu R, Cheng L, Cheng Q. Low-intensity pulsed ultrasound promotes mesenchymal stem cell transplantation-based articular cartilage regeneration via inhibiting the TNF signaling pathway. Stem Cell Res Ther 2023; 14:93. [PMID: 37069673 PMCID: PMC10111837 DOI: 10.1186/s13287-023-03296-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 03/22/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) transplantation therapy is highly investigated for the regenerative repair of cartilage defects. Low-intensity pulsed ultrasound (LIPUS) has the potential to promote chondrogenic differentiation of MSCs. However, its underlying mechanism remains unclear. Here, we investigated the promoting effects and mechanisms underlying LIPUS stimulation on the chondrogenic differentiation of human umbilical cord mesenchymal stem cells (hUC-MSCs) and further evaluated its regenerative application value in articular cartilage defects in rats. METHODS LIPUS was applied to stimulate cultured hUC-MSCs and C28/I2 cells in vitro. Immunofluorescence staining, qPCR analysis, and transcriptome sequencing were used to detect mature cartilage-related markers of gene and protein expression for a comprehensive evaluation of differentiation. Injured articular cartilage rat models were established for further hUC-MSC transplantation and LIPUS stimulation in vivo. Histopathology and H&E staining were used to evaluate the repair effects of the injured articular cartilage with LIPUS stimulation. RESULTS The results showed that LIPUS stimulation with specific parameters effectively promoted the expression of mature cartilage-related genes and proteins, inhibited TNF-α gene expression in hUC-MSCs, and exhibited anti-inflammation in C28/I2 cells. In addition, the articular cartilage defects of rats were significantly repaired after hUC-MSC transplantation and LIPUS stimulation. CONCLUSIONS Taken together, LIPUS stimulation could realize articular cartilage regeneration based on hUC-MSC transplantation due to the inhibition of the TNF signaling pathway, which is of clinical value for the relief of osteoarthritis.
Collapse
Affiliation(s)
- Yiming Chen
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Huiyi Yang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhaojie Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
- School of Life Science and Technology, Tongji University, Shanghai, 200065, China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
- School of Life Science and Technology, Tongji University, Shanghai, 200065, China
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Qian Cheng
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China.
- Frontiers Science Center for Intelligent Autonomous Systems, Shanghai, 201210, China.
| |
Collapse
|
15
|
Gupta D, Savva J, Li X, Chandler JH, Shelton RM, Scheven BA, Mulvana H, Valdastri P, Lucas M, Walmsley AD. Traditional Multiwell Plates and Petri Dishes Limit the Evaluation of the Effects of Ultrasound on Cells In Vitro. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1745-1761. [PMID: 35760602 DOI: 10.1016/j.ultrasmedbio.2022.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 06/15/2023]
Abstract
Ultrasound accelerates healing in fractured bone; however, the mechanisms responsible are poorly understood. Experimental setups and ultrasound exposures vary or are not adequately characterized across studies, resulting in inter-study variation and difficulty in concluding biological effects. This study investigated experimental variability introduced through the cell culture platform used. Continuous wave ultrasound (45 kHz; 10, 25 or 75 mW/cm2, 5 min/d) was applied, using a Duoson device, to Saos-2 cells seeded in multiwell plates or Petri dishes. Pressure field and vibration quantification and finite-element modelling suggested formation of complex interference patterns, resulting in localized displacement and velocity gradients, more pronounced in multiwell plates. Cell experiments revealed lower metabolic activities in both culture platforms at higher ultrasound intensities and absence of mineralization in certain regions of multiwell plates but not in Petri dishes. Thus, the same transducer produced variable results in different cell culture platforms. Analysis on Petri dishes further revealed that higher intensities reduced vinculin expression and distorted cell morphology, while causing mitochondrial and endoplasmic reticulum damage and accumulation of cells in sub-G1 phase, leading to cell death. More defined experimental setups and reproducible ultrasound exposure systems are required to study the real effect of ultrasound on cells for development of effective ultrasound-based therapies not just limited to bone repair and regeneration.
Collapse
Affiliation(s)
- Dhanak Gupta
- School of Dentistry, University of Birmingham, Birmingham, UK.
| | - Jill Savva
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | - Xuan Li
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | - James H Chandler
- Science and Technology of Robotics in Medicine (STORM) Laboratory UK, School of Electronic and Electrical Engineering, University of Leeds, Leeds, UK
| | | | - Ben A Scheven
- School of Dentistry, University of Birmingham, Birmingham, UK
| | - Helen Mulvana
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, UK
| | - Pietro Valdastri
- Science and Technology of Robotics in Medicine (STORM) Laboratory UK, School of Electronic and Electrical Engineering, University of Leeds, Leeds, UK
| | - Margaret Lucas
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
16
|
Fielder M, Nair AK. Bone tissue growth in ultrasonically stimulated bioinspired scaffolds. Comput Methods Biomech Biomed Engin 2022:1-6. [PMID: 35971823 DOI: 10.1080/10255842.2022.2109415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We develop computational models of bone growth in ultrasonically stimulated porous tissue scaffolds with uniform square pores and a bioinspired structure. While bone growth in the bioinspired scaffolds is slower, it produces amounts of bone comparable to the square pore scaffold, making the bioinspired structure ideal for enhancing bone growth with better structural integrity. Controlling the initial mesenchymal stem cell distribution in the scaffolds also affects the growth rate and total bone formation, which could be further useful for controlling bone growth in the scaffold based on an individual's physiology.
Collapse
Affiliation(s)
- Marco Fielder
- Multiscale Materials Modeling Lab, Department of Mechanical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Arun K Nair
- Multiscale Materials Modeling Lab, Department of Mechanical Engineering, University of Arkansas, Fayetteville, AR, USA.,Institute for Nanoscience and Engineering, University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|
17
|
Xia P, Shi Y, Wang X, Li X. Advances in the application of low-intensity pulsed ultrasound to mesenchymal stem cells. Stem Cell Res Ther 2022; 13:214. [PMID: 35619156 PMCID: PMC9137131 DOI: 10.1186/s13287-022-02887-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/03/2022] [Indexed: 11/10/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are stem cells that exhibit self-renewal capacity and multi-directional differentiation potential. They can be extracted from the bone marrow and umbilical cord, as well as adipose, amnion, and other tissues. They are widely used in tissue engineering and are currently considered an important source of cells in the field of regenerative medicine. Since certain limitations, such as an insufficient cell source, mature differentiation, and low transplantation efficiency, are still associated with MSCs, researchers have currently focused on improving the efficacy of MSCs. Low-intensity pulsed ultrasound (LIPUS) has mechanical, cavitation, and thermal effects that can produce different biological effects on organs, tissues, and cells. It can be used for fracture treatment, cartilage repair, and stem cell applications. An in-depth study of the role and mechanism of action of LIPUS in MSC treatment would promote our understanding of LIPUS and promote research in this field. In this article, we have reviewed the progress in research on the use of LIPUS with various MSCs and comprehensively discussed the progress in the use of LIPUS for promoting the proliferation, differentiation, and migration of MSCs, as well as its future prospects.
Collapse
Affiliation(s)
- Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Yi Shi
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaoju Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
18
|
Mechanosensitive Osteogenesis on Native Cellulose Scaffolds for Bone Tissue Engineering. J Biomech 2022; 135:111030. [DOI: 10.1016/j.jbiomech.2022.111030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 01/17/2022] [Accepted: 02/28/2022] [Indexed: 12/23/2022]
|
19
|
Hadaegh Y, Uludag H, Dederich D, El-Bialy TH. The effect of low intensity pulsed ultrasound on mandibular condylar growth in young adult rats. Bone Rep 2021; 15:101122. [PMID: 34527791 PMCID: PMC8433121 DOI: 10.1016/j.bonr.2021.101122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 11/30/2022] Open
Abstract
There is a need for more effective methods to enhance mandibular growth in young adults with mandibular deficiency. Previous studies suggest that low intensity pulsed ultrasound (LIPUS) can enhance mandibular growth in growing individuals. This study aimed to evaluate the potential growth changes of the mandible following 4-week LIPUS application in young adult rats. Nineteen ≈120-day-old female rats were allocated to experimental (n = 10) and control (n = 9) groups. The animals in the experimental group were treated with LIPUS to their temporomandibular joints (TMJs) bilaterally, 20 min each day for 28 consecutive days. Animals were then euthanized; gross morphological evaluation was performed on 2D photographs and 3D virtual models of hemi-mandibles, and microstructural assessment was done for the mandibular condyle (MC). Evaluation of mineralization and microarchitecture properties of subchondral cancellous bone was performed by micro-computed tomography (μCT) scanning. Qualitative and histomorphometric analysis was done on condylar cartilage and subchondral bone following Alcian Blue/PAS and Goldner's Trichrome staining. Vital flourochrome (calcein green) labeling was also utilized to determine the amount of endochondral bone growth. Gross morphological evaluations showed a slight statistically non-significant increase especially in the main condylar growth direction in the LIPUS group. Moreover, 3D evaluation depicted an enhanced periosteal bone apposition at the site of LIPUS application. Microstructural analysis revealed that LIPUS stimulates both chondrogenesis and osteogenesis and enhances endochondral bone formation in young adult rat MC. Furthermore, the effect of LIPUS on osteogenic cells of subchondral cancellous bone was notable. To conclude, LIPUS can enhance young adult rats' MC residual growth potential.
Collapse
Affiliation(s)
- Yasamin Hadaegh
- School of Dentistry, University of Alberta, Edmonton, Canada
| | - Hasan Uludag
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
20
|
Marsudi MA, Ariski RT, Wibowo A, Cooper G, Barlian A, Rachmantyo R, Bartolo PJDS. Conductive Polymeric-Based Electroactive Scaffolds for Tissue Engineering Applications: Current Progress and Challenges from Biomaterials and Manufacturing Perspectives. Int J Mol Sci 2021; 22:11543. [PMID: 34768972 PMCID: PMC8584045 DOI: 10.3390/ijms222111543] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
The practice of combining external stimulation therapy alongside stimuli-responsive bio-scaffolds has shown massive potential for tissue engineering applications. One promising example is the combination of electrical stimulation (ES) and electroactive scaffolds because ES could enhance cell adhesion and proliferation as well as modulating cellular specialization. Even though electroactive scaffolds have the potential to revolutionize the field of tissue engineering due to their ability to distribute ES directly to the target tissues, the development of effective electroactive scaffolds with specific properties remains a major issue in their practical uses. Conductive polymers (CPs) offer ease of modification that allows for tailoring the scaffold's various properties, making them an attractive option for conductive component in electroactive scaffolds. This review provides an up-to-date narrative of the progress of CPs-based electroactive scaffolds and the challenge of their use in various tissue engineering applications from biomaterials perspectives. The general issues with CP-based scaffolds relevant to its application as electroactive scaffolds were discussed, followed by a more specific discussion in their applications for specific tissues, including bone, nerve, skin, skeletal muscle and cardiac muscle scaffolds. Furthermore, this review also highlighted the importance of the manufacturing process relative to the scaffold's performance, with particular emphasis on additive manufacturing, and various strategies to overcome the CPs' limitations in the development of electroactive scaffolds.
Collapse
Affiliation(s)
- Maradhana Agung Marsudi
- Materials Science and Engineering Research Group, Faculty of Mechanical and Aerospace Engineering, Institut Teknologi Bandung, Jl. Ganesha 10, Bandung 40132, West Java, Indonesia; (M.A.M.); (R.T.A.); (R.R.)
| | - Ridhola Tri Ariski
- Materials Science and Engineering Research Group, Faculty of Mechanical and Aerospace Engineering, Institut Teknologi Bandung, Jl. Ganesha 10, Bandung 40132, West Java, Indonesia; (M.A.M.); (R.T.A.); (R.R.)
| | - Arie Wibowo
- Materials Science and Engineering Research Group, Faculty of Mechanical and Aerospace Engineering, Institut Teknologi Bandung, Jl. Ganesha 10, Bandung 40132, West Java, Indonesia; (M.A.M.); (R.T.A.); (R.R.)
- Research Center for Nanoscience and Nanotechnology, Institut Teknologi Bandung, Jl. Ganesha 10, Bandung 40132, West Java, Indonesia
| | - Glen Cooper
- Department of Mechanical, Aerospace, and Civil Engineering, University of Manchester, Manchester M13 9PL, UK; (G.C.); (P.J.D.S.B.)
| | - Anggraini Barlian
- School of Life Science & Technology, Institut Teknologi Bandung, Jl. Ganesha 10, Bandung 40132, West Java, Indonesia;
| | - Riska Rachmantyo
- Materials Science and Engineering Research Group, Faculty of Mechanical and Aerospace Engineering, Institut Teknologi Bandung, Jl. Ganesha 10, Bandung 40132, West Java, Indonesia; (M.A.M.); (R.T.A.); (R.R.)
| | - Paulo J. D. S. Bartolo
- Department of Mechanical, Aerospace, and Civil Engineering, University of Manchester, Manchester M13 9PL, UK; (G.C.); (P.J.D.S.B.)
| |
Collapse
|
21
|
Jin M, Seo SH, Kim BS, Hwang S, Kang YG, Shin JW, Cho KH, Byeon J, Shin MC, Kim D, Yoon C, Min KA. Combined Application of Prototype Ultrasound and BSA-Loaded PLGA Particles for Protein Delivery. Pharm Res 2021; 38:1455-1466. [PMID: 34398405 DOI: 10.1007/s11095-021-03091-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/02/2021] [Indexed: 01/22/2023]
Abstract
PURPOSE To develop an in vitro culture system for tissue engineering to mimic the in vivo environment and evaluate the applicability of ultrasound and PLGA particle system. METHODS For tissue engineering, large molecules such as growth factors for cell differentiation should be supplied in a controlled manner into the culture system, and the in vivo microenvironment need to be reproduced in the system for the regulation of cellular function. In this study, portable prototype ultrasound with low intensity was devised and tested for protein release from bovine serum albumin (BSA)-loaded poly(lactic-co-glycolic acid) (PLGA) particles. RESULTS BSA-loaded PLGA particles were prepared using various types of PLGA reagents and their physicochemical properties were characterized including particle size, shape, or aqueous wetting profiles. The BSA-loaded formulation showed nano-ranged size distribution with optimal physical stability during storage period, and protein release behaviors in a controlled manner. Notably, the application of prototype ultrasound with low intensity influenced protein release patterns in the culture system containing the BSA-loaded PLGA formulation. The results revealed that the portable ultrasound set controlled by the computer could contribute for the protein delivery in the culture medium. CONCLUSIONS This study suggests that combined application with ultrasound and protein-loaded PLGA encapsulation system could be utilized to improve culture system for tissue engineering or cell regeneration therapy.
Collapse
Affiliation(s)
| | | | - Bo Seok Kim
- Department of Nanoscience and Engineering, School of Biomedical Engineering, Inje University, Gimhae, 50834, Republic of Korea
| | - Seungmi Hwang
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Yun Gyeong Kang
- Department of Biomedical Engineering, Inje University, Gimhae, 50834, Republic of Korea
| | - Jung-Woog Shin
- Department of Biomedical Engineering, Inje University, Gimhae, 50834, Republic of Korea
| | - Kwan Hyung Cho
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Jimi Byeon
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Meong Cheol Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam, 52828, Republic of Korea
| | - Doyeon Kim
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae, Gyeongnam, 50834, Republic of Korea
| | - Changhan Yoon
- Department of Nanoscience and Engineering, School of Biomedical Engineering, Inje University, Gimhae, 50834, Republic of Korea. .,Department of Biomedical Engineering, Inje University, Gimhae, 50834, Republic of Korea.
| | - Kyoung Ah Min
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae, Gyeongnam, 50834, Republic of Korea.
| |
Collapse
|
22
|
Yang J, Tang Z, Liu Y, Luo Z, Xiao Y, Zhang X. Comparison of chondro-inductivity between collagen and hyaluronic acid hydrogel based on chemical/physical microenvironment. Int J Biol Macromol 2021; 182:1941-1952. [PMID: 34062160 DOI: 10.1016/j.ijbiomac.2021.05.188] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/14/2021] [Accepted: 05/27/2021] [Indexed: 02/09/2023]
Abstract
Achieving chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) successfully is crucial for cartilage regeneration. To date, various hydrogels with different chemical microenvironment have been used to modulate chondrogenic differentiation of BMSCs, especially collagen and hyaluronic acid hydrogel. However, the chondro-inductive ability of collagen and hyaluronic acid hydrogel has not been evaluated yet and the different chemical and physical microenvironment of these two hydrogels increase the difficulty of comparison. In this study, three different hydrogels based on collagen and hyaluronic acid (self-assembled collagen hydrogel (Col), self-assembled collagen hydrogel cross-linked with genipin (Cgp), and methacrylated hyaluronic acid hydrogel (HA)) were prepared and their chondro-inductive ability on the encapsulated BMSCs was evaluated. Col and Cgp have the same chemical composition and similar microstructure, but are different from HA, while Cgp and HA hydrogels have the same mechanical strength. It was found that chemical and physical microenvironments of the hydrogels combined to influence cell condensation. Thanks to cell condensation was more likely to occur in collagen hydrogels in the early stage, the cartilage-induced ability was in the order of Col > Cgp > HA. However, the severe shrinkage of Col and Cgp resulted in no enough space for cell proliferation within hydrogels in the later stage. In contrast, relatively stable physical microenvironment of HA helped to maintain continuous production of cartilage-related matrix in the later stage. Overall, these results revealed that the chondro-inductive ability of collagen and hyaluronic acid hydrogel with different chemical and physical microenvironment cannot be evaluated by a particular time period. However, it provided important information for optimization and design of the future hydrogels towards successful repair of articular cartilage.
Collapse
Affiliation(s)
- Jirong Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China; Research Center for Human Tissue and Organs Degeneration, Institute Biomedical and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangzhou, China
| | - Zizhao Tang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| | - Yifan Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| | - Zhaocong Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| | - Yumei Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| |
Collapse
|
23
|
Application of Computational Method in Designing a Unit Cell of Bone Tissue Engineering Scaffold: A Review. Polymers (Basel) 2021; 13:polym13101584. [PMID: 34069101 PMCID: PMC8156807 DOI: 10.3390/polym13101584] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/27/2022] Open
Abstract
The design of a scaffold of bone tissue engineering plays an important role in ensuring cell viability and cell growth. Therefore, it is a necessity to produce an ideal scaffold by predicting and simulating the properties of the scaffold. Hence, the computational method should be adopted since it has a huge potential to be used in the implementation of the scaffold of bone tissue engineering. To explore the field of computational method in the area of bone tissue engineering, this paper provides an overview of the usage of a computational method in designing a unit cell of bone tissue engineering scaffold. In order to design a unit cell of the scaffold, we discussed two categories of unit cells that can be used to design a feasible scaffold, which are non-parametric and parametric designs. These designs were later described and being categorised into multiple types according to their characteristics, such as circular structures and Triply Periodic Minimal Surface (TPMS) structures. The advantages and disadvantages of these designs were discussed. Moreover, this paper also represents some software that was used in simulating and designing the bone tissue scaffold. The challenges and future work recommendations had also been included in this paper.
Collapse
|
24
|
Wang P, Sun Y, Shi X, Shen H, Ning H, Liu H. Bioscaffolds embedded with regulatory modules for cell growth and tissue formation: A review. Bioact Mater 2021; 6:1283-1307. [PMID: 33251379 PMCID: PMC7662879 DOI: 10.1016/j.bioactmat.2020.10.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/07/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
The demand for artificial organs has greatly increased because of various aging-associated diseases and the wide need for organ transplants. A recent trend in tissue engineering is the precise reconstruction of tissues by the growth of cells adhering to bioscaffolds, which are three-dimensional (3D) structures that guide tissue and organ formation. Bioscaffolds used to fabricate bionic tissues should be able to not only guide cell growth but also regulate cell behaviors. Common regulation methods include biophysical and biochemical stimulations. Biophysical stimulation cues include matrix hardness, external stress and strain, surface topology, and electromagnetic field and concentration, whereas biochemical stimulation cues include growth factors, proteins, kinases, and magnetic nanoparticles. This review discusses bioink preparation, 3D bioprinting (including extrusion-based, inkjet, and ultraviolet-assisted 3D bioprinting), and regulation of cell behaviors. In particular, it provides an overview of state-of-the-art methods and devices for regulating cell growth and tissue formation and the effects of biophysical and biochemical stimulations on cell behaviors. In addition, the fabrication of bioscaffolds embedded with regulatory modules for biomimetic tissue preparation is explained. Finally, challenges in cell growth regulation and future research directions are presented.
Collapse
Affiliation(s)
- Pengju Wang
- Department of Mechanical Manufacturing and Automation, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Yazhou Sun
- Department of Mechanical Manufacturing and Automation, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Xiaoquan Shi
- Department of Mechanical Manufacturing and Automation, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Huixing Shen
- Department of Mechanical Manufacturing and Automation, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Haohao Ning
- Department of Mechanical Manufacturing and Automation, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Haitao Liu
- Department of Mechanical Manufacturing and Automation, School of Mechatronics Engineering, Harbin Institute of Technology, Harbin, 150001, China
| |
Collapse
|
25
|
Jin M, Kim BS, Seo SH, Kim M, Kang YG, Shin JW, Cho KH, Shin MC, Yoon C, Min KA. Synergistic Effect of Growth Factor Releasing Polymeric Nanoparticles and Ultrasound Stimulation on Osteogenic Differentiation. Pharmaceutics 2021; 13:pharmaceutics13040457. [PMID: 33801692 PMCID: PMC8066944 DOI: 10.3390/pharmaceutics13040457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 01/16/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been extensively used in the tissue regeneration therapy. Ex vivo therapy with well-differentiated osteogenic cells is known as an efficient treatment for musculoskeletal diseases, including rheumatoid diseases. However, along with its high cost, the current therapy has limitations in terms of restoring bone regeneration procedures. An efficient process for the cell differentiation to obtain a large number of functionalized osteogenic cells is necessary. Therefore, it is strongly recommended to develop strategies to produce sufficient numbers of well-differentiated osteogenic cells from the MSCs. In general, differentiation media with growth factors have been used to facilitate cell differentiation. In the present study, the poly (lactic-co-glycolic acid) (PLGA) nanoparticles incorporating the growth factors were included in the media, resulting in releasing growth factors (dexamethasone and β-glycerophosphate) in the media in the controlled manner. Stable growth and early differentiation of osteogenic cells were achieved by the PLGA-based growth factor releasing system. Moreover, low intensity pulsed ultrasound was applied to this system to induce cell differentiation process. The results revealed that, as a biomarker at early stage of osteogenic cell differentiation, Lamin A/C nuclear protein was efficiently expressed in the cells growing in the presence of PLGA-based growth factor reservoirs and ultrasound. In conclusion, our results showed that the ultrasound stimulation combined with polymeric nanoparticles releasing growth factors could potentially induce osteogenic cell differentiation.
Collapse
Affiliation(s)
- Minki Jin
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea; (M.J.); (K.H.C.)
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| | - Bo Seok Kim
- Department of Nanoscience and Engineering, School of Biomedical Engineering, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea; (B.S.K.); (S.H.S.)
| | - Sung Ho Seo
- Department of Nanoscience and Engineering, School of Biomedical Engineering, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea; (B.S.K.); (S.H.S.)
| | - Minjeong Kim
- Department of Biomedical Engineering, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea; (M.K.); (Y.G.K.); (J.-W.S.)
| | - Yun Gyeong Kang
- Department of Biomedical Engineering, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea; (M.K.); (Y.G.K.); (J.-W.S.)
| | - Jung-Woog Shin
- Department of Biomedical Engineering, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea; (M.K.); (Y.G.K.); (J.-W.S.)
| | - Kwan Hyung Cho
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea; (M.J.); (K.H.C.)
| | - Meong Cheol Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju 52828, Gyeongnam, Korea;
| | - Changhan Yoon
- Department of Nanoscience and Engineering, School of Biomedical Engineering, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea; (B.S.K.); (S.H.S.)
- Department of Biomedical Engineering, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea; (M.K.); (Y.G.K.); (J.-W.S.)
- Correspondence: (C.Y.); (K.A.M.); Tel.: +82-55-320-3301 (C.Y.); +82-55-320-3459 (K.A.M.)
| | - Kyoung Ah Min
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae 50834, Gyeongnam, Korea; (M.J.); (K.H.C.)
- Correspondence: (C.Y.); (K.A.M.); Tel.: +82-55-320-3301 (C.Y.); +82-55-320-3459 (K.A.M.)
| |
Collapse
|
26
|
Su X, Wang T, Guo S. Applications of 3D printed bone tissue engineering scaffolds in the stem cell field. Regen Ther 2021; 16:63-72. [PMID: 33598507 PMCID: PMC7868584 DOI: 10.1016/j.reth.2021.01.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/07/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
Due to traffic accidents, injuries, burns, congenital malformations and other reasons, a large number of patients with tissue or organ defects need urgent treatment every year. The shortage of donors, graft rejection and other problems cause a deficient supply for organ and tissue replacement, repair and regeneration of patients, so regenerative medicine came into being. Stem cell therapy plays an important role in the field of regenerative medicine, but it is difficult to fill large tissue defects by injection alone. The scientists combine three-dimensional (3D) printed bone tissue engineering scaffolds with stem cells to achieve the desired effect. These scaffolds can mimic the extracellular matrix (ECM), bone and cartilage, and eventually form functional tissues or organs by providing structural support and promoting attachment, proliferation and differentiation. This paper mainly discussed the applications of 3D printed bone tissue engineering scaffolds in stem cell regenerative medicine. The application examples of different 3D printing technologies and different raw materials are introduced and compared. Then we discuss the superiority of 3D printing technology over traditional methods, put forward some problems and limitations, and look forward to the future.
Collapse
Key Words
- 3D printing
- 3D, three-dimensional
- ABS, Acrylonitrile Butadiene Styrene plastic
- AM, additive manufacturing
- ASCs, adult stem cells
- Alg, alginate
- BCP, biphasic calcium phosphate
- BMSCs, bone marrow-derived mesenchymal stem cells
- Bone tissue engineering
- CAD, computer-aided design
- CAP, cold atmospheric plasma
- CHMA, chitosan methacrylate
- CT, computed tomography
- DCM, dichloromethane
- ECM, extracellular matrix
- ESCs, embryonic stem cells
- FDM, fused deposition molding
- GO, graphene oxide
- HA, hydroxyapatite
- HAp, hydroxyapatite nanoparticles
- HTy, 4-hydroxyphenethyl 2-(4-hydroxyphenyl) acetate
- LDM, Low Temperature Deposition Modeling
- LIPUS, low intensity pulsed ultrasound
- MBG/SA–SA, mesoporous bioactive glass/sodium alginate-sodium alginate
- MSCs, Marrow stem cells
- PC, Polycarbonate
- PCL, polycraprolactone
- PDA, polydopamine
- PED, Precision Extrusion Deposition
- PEG, Polyethylene glycol
- PEGDA, poly (ethylene glycol) diacrylate
- PLGA, poly (lactide-co-glycolide)
- PLLA, poly l-lactide
- PPSU, Polyphenylene sulfone resins
- PRF, platelet-rich fibrin
- PVA, polyvinyl alcohol
- RAD16-I, a soft nanofibrous self-assembling peptide
- SCAPs, human stem cells from the apical papilla
- SF-BG, silk fibroin and silk fibroin-bioactive glass
- SLA, Stereolithography
- SLM, Selective Laser Melting
- STL, standard tessellation language
- Scaffold materials
- Stem cells
- TCP, β-tricalcium phosphate
- dECM, decellularized bovine cartilage extracellular matrix
- hADSC, human adipose derived stem cells
- hMSCs, human mesenchymal stem cells
- iPS, induced pluripotent stem
- pcHμPs, novel self-healable pre-cross- linked hydrogel microparticles
- rBMSCs, rat bone marrow stem cells
Collapse
Affiliation(s)
- Xin Su
- Department of Plastic Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang 110001, Liaoning, People's Republic of China
| | - Ting Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang 110001, Liaoning, People's Republic of China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Shenyang 110001, Liaoning, People's Republic of China
| |
Collapse
|
27
|
Herreros-Pomares A, Zhou X, Calabuig-Fariñas S, Lee SJ, Torres S, Esworthy T, Hann SY, Jantus-Lewintre E, Camps C, Zhang LG. 3D printing novel in vitro cancer cell culture model systems for lung cancer stem cell study. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111914. [PMID: 33641907 DOI: 10.1016/j.msec.2021.111914] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/20/2021] [Accepted: 01/23/2021] [Indexed: 12/21/2022]
Abstract
Two-dimensional (2D) in vitro cell cultures and laboratory animals have been used traditionally as the gold-standard preclinical cancer model systems. However, for cancer stem cell (CSC) studies, they exhibit notable limitations on simulating native environment, which depreciate their translatability for clinical development purposes. In this study, different three-dimensional (3D) printing platforms were used to establish novel 3D cell cultures enriched in CSCs from non-small cell lung cancer (NSCLC) patients and cell lines. Rigid scaffolds with an elevated compressive modulus and uniform pores and channels were produced using different filaments. Hydrogel-based scaffolds were printed with a more irregular distribution of pores and a lower compressive modulus. As a 3D model of reference, suspension spheroid cultures were established. Therein, cancer cell lines exhibited enhanced proliferation profiles on rigid scaffolds compared to the same cells grown on either hydrogel scaffolds or tumor spheres. Meanwhile, primary cancer cells grew considerably better on hydrogel scaffolds or in tumor sphere culture, compared to cells grown on rigid scaffolds. Gene expression analysis confirmed that tumor spheres and cells seeded on hydrogel scaffolds significantly overexpress most of stemness and invasion promoters tested compared to control cells grown in 2D culture. A different phenomenon was observed within cells growing on the rigid scaffolds, where fewer significant variations in gene expression were detected. Our findings provide strong evidence for the advantageous usage of 3D printed models, especially those which use GelMA-PEGDA hydrogels as the primary scaffold material, for studying lung CSCs. The results demonstrated that the 3D printed scaffolds were better to mimic tumor complexity and regulate cancer cell behavior than in vivo 2D culture models.
Collapse
Affiliation(s)
- Alejandro Herreros-Pomares
- Mixed Unit TRIAL, Fundación Investigacíón Hospital General Universitario de Valencia & Centro de Investigación Príncipe Felipe, Valencia, Spain; CIBERONC, Valencia, Spain
| | - Xuan Zhou
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, United States
| | - Silvia Calabuig-Fariñas
- Mixed Unit TRIAL, Fundación Investigacíón Hospital General Universitario de Valencia & Centro de Investigación Príncipe Felipe, Valencia, Spain; CIBERONC, Valencia, Spain; Department of Pathology, Universitat de València, Valencia, Spain
| | - Se-Jun Lee
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, United States
| | - Susana Torres
- Mixed Unit TRIAL, Fundación Investigacíón Hospital General Universitario de Valencia & Centro de Investigación Príncipe Felipe, Valencia, Spain; CIBERONC, Valencia, Spain
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, United States
| | - Sung Yun Hann
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, United States
| | - Eloísa Jantus-Lewintre
- Mixed Unit TRIAL, Fundación Investigacíón Hospital General Universitario de Valencia & Centro de Investigación Príncipe Felipe, Valencia, Spain; CIBERONC, Valencia, Spain; Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| | - Carlos Camps
- Mixed Unit TRIAL, Fundación Investigacíón Hospital General Universitario de Valencia & Centro de Investigación Príncipe Felipe, Valencia, Spain; CIBERONC, Valencia, Spain; Department of Medical Oncology, Hospital General Universitario de Valencia, Valencia, Spain; Department of Medicine, Universitat de València, Valencia, Spain.
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, United States; Department of Biomedical Engineering, The George Washington University, Washington, DC, United States; Department of Electrical and Computer Engineering, The George Washington University, Washington, DC, United States; Department of Medicine, The George Washington University, Washington, DC, United States.
| |
Collapse
|
28
|
Chen M, Li Y, Liu S, Feng Z, Wang H, Yang D, Guo W, Yuan Z, Gao S, Zhang Y, Zha K, Huang B, Wei F, Sang X, Tian Q, Yang X, sui X, Zhou Y, Zheng Y, Guo Q. Hierarchical macro-microporous WPU-ECM scaffolds combined with Microfracture Promote in Situ Articular Cartilage Regeneration in Rabbits. Bioact Mater 2020; 6:1932-1944. [PMID: 33426368 PMCID: PMC7772526 DOI: 10.1016/j.bioactmat.2020.12.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/19/2020] [Accepted: 12/05/2020] [Indexed: 12/20/2022] Open
Abstract
Tissue engineering provides a promising avenue for treating cartilage defects. However, great challenges remain in the development of structurally and functionally optimized scaffolds for cartilage repair and regeneration. In this study, decellularized cartilage extracellular matrix (ECM) and waterborne polyurethane (WPU) were employed to construct WPU and WPU-ECM scaffolds by water-based 3D printing using low-temperature deposition manufacturing (LDM) system, which combines rapid deposition manufacturing with phase separation techniques. The scaffolds successfully achieved hierarchical macro‐microporous structures. After adding ECM, WPU scaffolds were markedly optimized in terms of porosity, hydrophilia and bioactive components. Moreover, the optimized WPU-ECM scaffolds were found to be more suitable for cell distribution, adhesion, and proliferation than the WPU scaffolds. Most importantly, the WPU-ECM scaffold could facilitate the production of glycosaminoglycan (GAG) and collagen and the upregulation of cartilage-specific genes. These results indicated that the WPU-ECM scaffold with hierarchical macro‐microporous structures could recreate a favorable microenvironment for cell adhesion, proliferation, differentiation, and ECM production. In vivo studies further revealed that the hierarchical macro‐microporous WPU-ECM scaffold combined with the microfracture procedure successfully regenerated hyaline cartilage in a rabbit model. Six months after implantation, the repaired cartilage showed a similar histological structure and mechanical performance to that of normal cartilage. In conclusion, the hierarchical macro‐microporous WPU-ECM scaffold may be a promising candidate for cartilage tissue engineering applications in the future.
Hierarchical macro‐microporous scaffolds could be fabricated by low-temperature deposition manufacturing. Waterborne polyurethane (WPU) scaffolds were optimized by adding decellularized cartilage extracellular matrix (ECM). The WPU-ECM scaffold provided a suitable microenvironment for cell attachment, proliferation, and differentiation in vitro. The paradigm of WPU-ECM scaffold and microfracture (MF) has great potential for clinical application in cartilage repair.
Collapse
Affiliation(s)
- Mingxue Chen
- Department of Orthopaedic Surgery, Peking University Fourth School of Clinical Medicine, Beijing Jishuitan Hospital, No. 31 Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - YangYang Li
- Academy for Advanced Interdisciplinary Studies, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, People's Republic of China
| | - Shuyun Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Zhaoxuan Feng
- School of Material Science and Engineering, University of Science and Technology Beijing, No. 30 Xueyuan Road, Haidian District, Beijing, 100083, People's Republic of China
| | - Hao Wang
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Road, Luzhou, 646000, People's Republic of China
| | - Dejin Yang
- Department of Orthopaedic Surgery, Peking University Fourth School of Clinical Medicine, Beijing Jishuitan Hospital, No. 31 Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
| | - Weimin Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Zhiguo Yuan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Shuang Gao
- Academy for Advanced Interdisciplinary Studies, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, People's Republic of China
| | - Yu Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Kangkang Zha
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Bo Huang
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Road, Luzhou, 646000, People's Republic of China
| | - Fu Wei
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Xinyu Sang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Qinyu Tian
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Xuan Yang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Xiang sui
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Yixin Zhou
- Department of Orthopaedic Surgery, Peking University Fourth School of Clinical Medicine, Beijing Jishuitan Hospital, No. 31 Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
- Corresponding author.
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, China
- Corresponding author.
| | - Quanyi Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
- Corresponding author.
| |
Collapse
|
29
|
Culibrk RA, Hahn MS. The Role of Chronic Inflammatory Bone and Joint Disorders in the Pathogenesis and Progression of Alzheimer's Disease. Front Aging Neurosci 2020; 12:583884. [PMID: 33364931 PMCID: PMC7750365 DOI: 10.3389/fnagi.2020.583884] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Late-onset Alzheimer's Disease (LOAD) is a devastating neurodegenerative disorder that causes significant cognitive debilitation in tens of millions of patients worldwide. Throughout disease progression, abnormal secretase activity results in the aberrant cleavage and subsequent aggregation of neurotoxic Aβ plaques in the cerebral extracellular space and hyperphosphorylation and destabilization of structural tau proteins surrounding neuronal microtubules. Both pathologies ultimately incite the propagation of a disease-associated subset of microglia-the principle immune cells of the brain-characterized by preferentially pro-inflammatory cytokine secretion and inhibited AD substrate uptake capacity, which further contribute to neuronal degeneration. For decades, chronic neuroinflammation has been identified as one of the cardinal pathophysiological driving features of AD; however, despite a number of works postulating the underlying mechanisms of inflammation-mediated neurodegeneration, its pathogenesis and relation to the inception of cognitive impairment remain obscure. Moreover, the limited clinical success of treatments targeting specific pathological features in the central nervous system (CNS) illustrates the need to investigate alternative, more holistic approaches for ameliorating AD outcomes. Accumulating evidence suggests significant interplay between peripheral immune activity and blood-brain barrier permeability, microglial activation and proliferation, and AD-related cognitive decline. In this work, we review a narrow but significant subset of chronic peripheral inflammatory conditions, describe how these pathologies are associated with the preponderance of neuroinflammation, and posit that we may exploit peripheral immune processes to design interventional, preventative therapies for LOAD. We then provide a comprehensive overview of notable treatment paradigms that have demonstrated considerable merit toward treating these disorders.
Collapse
Affiliation(s)
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| |
Collapse
|
30
|
Chansoria P, Schuchard K, Shirwaiker RA. Process hybridization schemes for multiscale engineered tissue biofabrication. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1673. [PMID: 33084240 DOI: 10.1002/wnan.1673] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/18/2022]
Abstract
Recapitulation of multiscale structure-function properties of cells, cell-secreted extracellular matrix, and 3D architecture of natural tissues is central to engineering biomimetic tissue substitutes. Toward achieving biomimicry, a variety of biofabrication processes have been developed, which can be broadly classified into five categories-fiber and fabric formation, additive manufacturing, surface modification, remote fields, and other notable processes-each with specific advantages and limitations. The majority of biofabrication literature has focused on using a single process at a time, which often limits the range of tissues that could be created with relevant features that span nano to macro scales. With multiscale biomimicry as the goal, development of hybrid biofabrication strategies that synergistically unite two or more processes to complement each other's strengths and limitations has been steadily increasing. This work discusses recent literature in this domain and attempts to equip the reader with the understanding of selecting appropriate processes that can harmonize toward creating engineered tissues with appropriate multiscale structure-function properties. Opportunities related to various hybridization schemes and a future outlook on scale-up biofabrication have also been discussed. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.
Collapse
Affiliation(s)
- Parth Chansoria
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Karl Schuchard
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Rohan A Shirwaiker
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA.,Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina, USA
| |
Collapse
|
31
|
Katiyar A, Osborn J, DasBanerjee M, Zhang LG, Sarkar K, Sarker KP. Inhibition of Human Breast Cancer Cell Proliferation by Low-Intensity Ultrasound Stimulation. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2020; 39:2043-2052. [PMID: 32352188 DOI: 10.1002/jum.15312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/17/2020] [Accepted: 04/03/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVES Cancer is characterized by uncontrolled cell proliferation, which makes novel therapies highly desired. In this study, the effects of near-field low-intensity pulsed ultrasound (LIPUS) stimulation on T47D human breast cancer cell and healthy immortalized MCF-12A breast epithelial cell proliferation were investigated in monolayer cultures. METHODS A customized ultrasound (US) exposure setup was used for the variation of key US parameters: intensity, excitation duration, and duty cycle. Cell proliferation was quantified by 5-bromo-2'-deoxyuridine and alamarBlue assays after LIPUS excitation. RESULTS At a 20% duty cycle and 10-minute excitation period, we varied LIPUS intensity from to 100 mW/cm2 (spatial-average temporal-average) to find a gradual decrease in T47D cell proliferation, the decrease being strongest at 100 mW/cm2 . In contrast, healthy MCF-12A breast cells showed an increase in proliferation when exposed to the same conditions. Above a 60% duty cycle, T47D cell proliferation decreased drastically. Effects of continuous wave US stimulation were further explored by varying the intensity and excitation period. CONCLUSIONS These experiments concluded that, irrespective of the waveform (pulsed or continuous), LIPUS stimulation could inhibit the proliferation of T47D breast cancer cells, whereas the same behavior was not observed in healthy cells. The study demonstrates the beneficial bioeffects of LIPUS on breast cancer cells and offers the possibility of developing novel US-mediated cancer therapy.
Collapse
Affiliation(s)
- Amit Katiyar
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| | - Jenna Osborn
- Department of Mechanical and Aerospace Engineering, George Washington University, Washington, DC, USA
| | - Malaya DasBanerjee
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, George Washington University, Washington, DC, USA
| | - Kausik Sarkar
- Department of Mechanical and Aerospace Engineering, George Washington University, Washington, DC, USA
| | - Krishna Pada Sarker
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
32
|
de Lucas B, Pérez LM, Bernal A, Gálvez BG. Ultrasound Therapy: Experiences and Perspectives for Regenerative Medicine. Genes (Basel) 2020; 11:genes11091086. [PMID: 32957737 PMCID: PMC7563547 DOI: 10.3390/genes11091086] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Ultrasound has emerged as a novel tool for clinical applications, particularly in the context of regenerative medicine. Due to its unique physico-mechanical properties, low-intensity ultrasound (LIUS) has been approved for accelerated fracture healing and for the treatment of established non-union, but its utility has extended beyond tissue engineering to other fields, including cell regeneration. Cells and tissues respond to acoustic ultrasound by switching on genetic repair circuits, triggering a cascade of molecular signals that promote cell proliferation, adhesion, migration, differentiation, and extracellular matrix production. LIUS also induces angiogenesis and tissue regeneration and has anti-inflammatory and anti-degenerative effects. Accordingly, the potential application of ultrasound for tissue repair/regeneration has been tested in several studies as a stand-alone treatment and, more recently, as an adjunct to cell-based therapies. For example, ultrasound has been proposed to improve stem cell homing to target tissues due to its ability to create a transitional and local gradient of cytokines and chemokines. In this review, we provide an overview of the many applications of ultrasound in clinical medicine, with a focus on its value as an adjunct to cell-based interventions. Finally, we discuss the various preclinical and clinical studies that have investigated the potential of ultrasound for regenerative medicine.
Collapse
Affiliation(s)
- Beatriz de Lucas
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (B.d.L.); (L.M.P.)
| | - Laura M. Pérez
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (B.d.L.); (L.M.P.)
| | - Aurora Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain;
| | - Beatriz G. Gálvez
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (B.d.L.); (L.M.P.)
- Correspondence:
| |
Collapse
|
33
|
Liu DD, Ullah M, Concepcion W, Dahl JJ, Thakor AS. The role of ultrasound in enhancing mesenchymal stromal cell-based therapies. Stem Cells Transl Med 2020; 9:850-866. [PMID: 32157802 PMCID: PMC7381806 DOI: 10.1002/sctm.19-0391] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 02/17/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been a popular platform for cell-based therapy in regenerative medicine due to their propensity to home to damaged tissue and act as a repository of regenerative molecules that can promote tissue repair and exert immunomodulatory effects. Accordingly, a great deal of research has gone into optimizing MSC homing and increasing their secretion of therapeutic molecules. A variety of methods have been used to these ends, but one emerging technique gaining significant interest is the use of ultrasound. Sound waves exert mechanical pressure on cells, activating mechano-transduction pathways and altering gene expression. Ultrasound has been applied both to cultured MSCs to modulate self-renewal and differentiation, and to tissues-of-interest to make them a more attractive target for MSC homing. Here, we review the various applications of ultrasound to MSC-based therapies, including low-intensity pulsed ultrasound, pulsed focused ultrasound, and extracorporeal shockwave therapy, as well as the use of adjunctive therapies such as microbubbles. At a molecular level, it seems that ultrasound transiently generates a local gradient of cytokines, growth factors, and adhesion molecules that facilitate MSC homing. However, the molecular mechanisms underlying these methods are far from fully elucidated and may differ depending on the ultrasound parameters. We thus put forth minimal criteria for ultrasound parameter reporting, in order to ensure reproducibility of studies in the field. A deeper understanding of these mechanisms will enhance our ability to optimize this promising therapy to assist MSC-based approaches in regenerative medicine.
Collapse
Affiliation(s)
- Daniel D. Liu
- Interventional Regenerative Medicine and Imaging Laboratory, Department of RadiologyStanford UniversityPalo AltoCalifornia
| | - Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Department of RadiologyStanford UniversityPalo AltoCalifornia
| | | | - Jeremy J. Dahl
- Interventional Regenerative Medicine and Imaging Laboratory, Department of RadiologyStanford UniversityPalo AltoCalifornia
| | - Avnesh S. Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of RadiologyStanford UniversityPalo AltoCalifornia
| |
Collapse
|
34
|
Zhou X, Tenaglio S, Esworthy T, Hann SY, Cui H, Webster TJ, Fenniri H, Zhang LG. Three-Dimensional Printing Biologically Inspired DNA-Based Gradient Scaffolds for Cartilage Tissue Regeneration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:33219-33228. [PMID: 32603082 DOI: 10.1021/acsami.0c07918] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cartilage damage caused by aging, repeated overloading, trauma, and diseases can result in chronic pain, inflammation, stiffness, and even disability. Unlike other types of tissues (bone, skin, muscle, etc.), cartilage tissue has an extremely weak regenerative capacity. Currently, the gold standard surgical treatment for repairing cartilage damage includes autografts and allografts. However, these procedures are limited by insufficient donor sources and the potential for immunological rejection. After years of development, engineered tissue now provides a valuable artificial replacement for tissue regeneration purposes. Three-dimensional (3D) bioprinting technologies can print customizable hierarchical structures with cells. The objective of the current work was to prepare a 3D-printed three-layer gradient scaffold with lysine-functionalized rosette nanotubes (RNTK) for improving the chondrogenic differentiation of adipose-derived mesenchymal stem cells (ADSCs). Specifically, biologically inspired RNTKs were utilized in our work because they have unique surface chemistry and biomimetic nanostructure to improve cell adhesion and growth. Different ratios of gelatin methacrylate (GelMA) and poly(ethylene glycol) diacrylate (PEGDA) were printed into a three-layer GelMA-PEGDA gradient scaffold using a stereolithography-based printer, followed by coating with RNTKs. The pores and channels (∼500 μm) were observed in the scaffold. It was found that the population of ADSCs on the GelMA-PEGDA-RNTK scaffold increased by 34% compared to the GelMA-PEGDA scaffold (control). Moreover, after 3 weeks of chondrogenic differentiation, collagen II, glycosaminoglycan, and total collagen synthesis on the GelMA-PEGDA-RNTK scaffold significantly respectively increased by 59%, 71%, and 60%, as compared to the control scaffold. Gene expression of collagen II α1, SOX 9, and aggrecan in the ADSCs growing on the GelMA-PEGDA-RNTK scaffold increased by 79%, 52%, and 47% after 3 weeks, compared to the controls, respectively. These results indicated that RNTKs are a promising type of nanotubes for promoting chondrogenic differentiation, and the present 3D-printed three-layer gradient GelMA-PEGDA-RNTK scaffold shows considerable promise for future cartilage repair and regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Hicham Fenniri
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | | |
Collapse
|
35
|
Minto J, Zhou X, Osborn J, Zhang LG, Sarkar K, Rao RD. Three-Dimensional Printing: A Catalyst for a Changing Orthopaedic Landscape. JBJS Rev 2020; 8:e0076. [DOI: 10.2106/jbjs.rvw.19.00076] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
36
|
Amini A, Chien S, Bayat M. Impact of Ultrasound Therapy on Stem Cell Differentiation - A Systematic Review. Curr Stem Cell Res Ther 2020; 15:462-472. [PMID: 32096749 DOI: 10.2174/1574888x15666200225124934] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This is a systematic review of the effects of low-intensity pulsed ultrasound (LIPUS) on stem cell differentiation. BACKGROUND DATA Recent studies have investigated several types of stem cells from different sources in the body. These stem cells should strictly be certified and promoted for cell therapies before being used in medical applications. LIPUS has been used extensively in treatment centers and in research to promote stem cell differentiation, function, and proliferation. MATERIALS AND METHODS The databases of PubMed, Google Scholar, and Scopus were searched for abstracts and full-text scientific papers published from 1989-2019 that reported the application of LIPUS on stem cell differentiation. Related English language articles were found using the following defined keywords: low-intensity pulsed ultrasound, stem cell, differentiation. Criteria for inclusion in the review were: LIPUS with frequencies of 1-3 MHz and pulsed ultrasound intensity of <500 mW/cm2. Duration, exposure time, and cell sources were taken into consideration. RESULTS Fifty-two articles were selected based on the inclusion criteria. Most articles demonstrated that the application of LIPUS had positive effects on stem cell differentiation. However, some authors recommended that LIPUS combined with other physical therapy aides was more effective in stem cell differentiation. CONCLUSION LIPUS significantly increases the level of stem cell differentiation in cells derived mainly from bone marrow mesenchymal stem cells. There is a need for further studies to analyze the effect of LIPUS on cells derived from other sources, particularly adipose tissue-derived mesenchymal stem cells, for treating hard diseases, such as osteoporosis and diabetic foot ulcer. Due to a lack of reporting on standard LIPUS parameters in the field, more experiments comparing the protocols for standardization of LIPUS parameters are needed to establish the best protocol, which would allow for the best results.
Collapse
Affiliation(s)
- Abdollah Amini
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville, Louisville, KY, United States
| | - Mohammad Bayat
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Price Institute of Surgical Research, University of Louisville, Louisville, KY, United States
| |
Collapse
|
37
|
Liu S, Zhou M, Li J, Hu B, Jiang D, Huang H, Song J. LIPUS inhibited the expression of inflammatory factors and promoted the osteogenic differentiation capacity of hPDLCs by inhibiting the NF-κB signaling pathway. J Periodontal Res 2019; 55:125-140. [PMID: 31541455 DOI: 10.1111/jre.12696] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 07/16/2019] [Accepted: 08/13/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVES As a chronic infectious disease, periodontitis could lead to tooth and bone loss. Low-intensity pulsed ultrasound (LIPUS) is a safe, noninvasive treatment method to effectively inhibit inflammation and promote bone differentiation. However, the application of LIPUS in curing periodontitis is still rare. Our study aimed to explore the ability of LIPUS to inhibit inflammatory factors and promote the osteogenic differentiation capacity of human periodontal ligament cells (hPDLCs), and its underlying mechanism. MATERIAL AND METHODS Human periodontal ligament cells were obtained and cultured from the premolar tissue samples for experiments. First, hPDLCs were treated for 24 hours using lipopolysaccharide (LPS) and then exposed to LIPUS (10 mW/cm2 , 30 mW/cm2 , 60 mW/cm2 , and 90 mW/cm2 ) to determine the appropriate intensity to inhibit expression of the inflammatory factors interleukin-6 (IL-6) and interleukin-8 (IL-8) expression. The expression of IL-6 and IL-8 was detected by real-time PCR and enzyme-linked immunosorbent assay. The safety of the most appropriate intensity of LIPUS was tested by a cell counting kit 8 test and an apoptosis assay. Then, LPS-induced hPDLCs were treated in osteogenic medium for 7-21 days with or without LIPUS (90 mW/cm2 , 30 min/d) stimulation. The osteogenic genes RUNX2, OPN, OSX, and OCN were measured by real-time PCR. Additionally, osteogenic differentiation capacity was determined using alkaline phosphatase (ALP) staining, ALP activity analysis, and Alizarin red staining. The activity of the nuclear factor-kappa B (NF-κB) signaling pathway was determined by western blotting, real-time PCR, immunofluorescence, and pathway blockade assays. RESULTS Lipopolysaccharide significantly upregulated the production and gene expression of IL-6 and IL-8, while LIPUS stimulation significantly inhibited IL-6 and IL-8 expression in an intensity-dependent manner. LIPUS (90 mW/cm2 ) was chosen as the most appropriate intensity, and there was no detrimental influence on cell proliferation and status with or without osteogenic medium. In addition, consecutive stimulation with LIPUS (90 mW/cm2 ) for 30 min/d for 7 days could also inhibit IL-6 and IL-8 gene expression, upregulate the expression of the osteogenesis-related genes RUNX2, OPN, OSX, and OCN, and promote osteogenic differentiation capacity in osteogenic medium in inflamed hPDLCs. The NF-κB signaling pathway was inhibited with LIPUS (90 mW/cm2 ) via inhibition of the phosphorylation of IκBα and the translocation of p65 into the nucleus in inflamed hPDLCs. Additional investigations of the NF-κB inhibitor, BAY 11-7082, revealed that LIPUS (90 mW/cm2 ) acted similarly to BAY 11-7802 to inhibit the NF-κB signaling pathway and increase osteogenesis-related genes and promote the osteogenic differentiation capacity of inflamed hPDLCs. CONCLUSION Low-intensity pulsed ultrasound (90 mW/cm2 ) stimulation could be a safe method to inhibit IL-6 and IL-8 in hPDLCs by inhibiting the NF-κB signaling pathway. The effect of LIPUS (90 mW/cm2 ) and BAY 11-7082 on LPS-induced inflammation demonstrated that both of these agents were capable of promoting osteogenesis-related gene expression and osteogenic differentiation in hPDLCs, suggesting that the effect of LIPUS on the promotion of osteogenic activity could be mediated in part through its ability to inhibit the NF-κB signal pathway. Hence, LIPUS could be a potential therapeutic method to cure periodontitis.
Collapse
Affiliation(s)
- Shan Liu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Mengjiao Zhou
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Jie Li
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Bo Hu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Dan Jiang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Hong Huang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Jinlin Song
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, College of Stomatology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
38
|
Yang J, Li Y, Liu Y, Li D, Zhang L, Wang Q, Xiao Y, Zhang X. Influence of hydrogel network microstructures on mesenchymal stem cell chondrogenesis in vitro and in vivo. Acta Biomater 2019; 91:159-172. [PMID: 31055122 DOI: 10.1016/j.actbio.2019.04.054] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/21/2019] [Accepted: 04/24/2019] [Indexed: 12/27/2022]
Abstract
Hydrogels, which provide three-dimensional (3D) niches for encapsulating bone marrow mesenchymal stem cells (BMSCs), are becoming a promising tissue engineering solution for chondrogenic differentiation of BMSCs. However, it remains a challenge to design a hydrogel material for effective chondrogenesis of BMSCs because of the complexity of cartilage ECM and cell-matrix interactions. Thus far, various studies have shown the physical-chemical cues of hydrogel materials to impact BMSCs chondrogenesis, but the design of the 3D network microstructure of the hydrogel to induce BMSCs chondrogenesis is still far from optimized. In this study, we successfully prepared two types of collagen hydrogels, namely, the fibrous network and porous network, with the same chemical composition and similar mechanical strength but with two distinct network microstructures. The two different network microstructures significantly influenced mass transfer, protein adsorption, degradability, and contraction of the collagen hydrogels. Moreover, the cells presented distinct proliferation and morphology in the two hydrogels, which consequently modulated chondrogenic differentiation of BMSCs derived from rat. Collagen hydrogels with a fibrous network promoted more chondrogenic differentiation of BMSCs without additional growth factors in vitro and subcutaneous implantation in vivo than those with a porous network. Moreover, fibrous network resulted in less ECM calcification than porous network. However, the fibrous network could not prevent hypertrophy of the chondrogenic cells induced by BMSCs. Overall, these results revealed that the 3D network microstructure of a hydrogel was a key design parameter for the chondrogenic differentiation of BMSCs. STATEMENT OF SIGNIFICANCE: Hydrogels had been used to induce the chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in cartilage tissue engineering, but the key design parameters remain unoptimized. This was mainly due to the different material properties including composition, strength, and microstructure, which would interplay with each other and result in difficulties to investigate the effects for one factor. In this study, we fabricated two collagen hydrogels with the same chemical composition and mechanical strength, but two distinct network microstructures. The effects of the two network microstructures on the chondrogenic differentiation of BMSCs were investigated by in vitro and in vivo assays. The results highlight the effects of network microstructures and provide important information about optimizing the design of future hydrogels in cartilage tissue engineering.
Collapse
Affiliation(s)
- Jirong Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| | - Yuanqi Li
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yanbo Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| | - Dongxiao Li
- Sichuan Academy of Chinese Medicine Science, Chengdu 61004, Sichuan, China
| | - Lei Zhang
- Sichuan Academy of Chinese Medicine Science, Chengdu 61004, Sichuan, China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China.
| | - Yumei Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| |
Collapse
|
39
|
Przekora A. Current Trends in Fabrication of Biomaterials for Bone and Cartilage Regeneration: Materials Modifications and Biophysical Stimulations. Int J Mol Sci 2019; 20:E435. [PMID: 30669519 PMCID: PMC6359292 DOI: 10.3390/ijms20020435] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 12/22/2022] Open
Abstract
The aim of engineering of biomaterials is to fabricate implantable biocompatible scaffold that would accelerate regeneration of the tissue and ideally protect the wound against biodevice-related infections, which may cause prolonged inflammation and biomaterial failure. To obtain antimicrobial and highly biocompatible scaffolds promoting cell adhesion and growth, materials scientists are still searching for novel modifications of biomaterials. This review presents current trends in the field of engineering of biomaterials concerning application of various modifications and biophysical stimulation of scaffolds to obtain implants allowing for fast regeneration process of bone and cartilage as well as providing long-lasting antimicrobial protection at the site of injury. The article describes metal ion and plasma modifications of biomaterials as well as post-surgery external stimulations of implants with ultrasound and magnetic field, providing accelerated regeneration process. Finally, the review summarizes recent findings concerning the use of piezoelectric biomaterials in regenerative medicine.
Collapse
Affiliation(s)
- Agata Przekora
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, W. Chodzki 1 Street, 20-093 Lublin, Poland.
| |
Collapse
|
40
|
Przekora A. The summary of the most important cell-biomaterial interactions that need to be considered during in vitro biocompatibility testing of bone scaffolds for tissue engineering applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 97:1036-1051. [PMID: 30678895 DOI: 10.1016/j.msec.2019.01.061] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 12/17/2022]
Abstract
Tissue engineered products (TEPs), which mean biomaterials containing either cells or growth factors or both cells and growth factors, may be used as an alternative to the autografts taken directly from the bone of the patients. Nevertheless, the use of TEPs needs much more understanding of biointeractions between biomaterials and eukaryotic cells. Despite the possibility of the use of in vitro cellular models for initial evaluation of the host response to the implanted biomaterial, it is observed that most researchers use cell cultures only for the evaluation of cytotoxicity and cell proliferation on the biomaterial surface, and then they proceed to animal models and in vivo testing of bone implants without fully utilizing the scientific potential of in vitro models. In this review, the most important biointeractions between eukaryotic cells and biomaterials were discussed, indicating molecular mechanisms of cell adhesion, proliferation, and biomaterial-induced activation of immune cells. The article also describes types of cellular models which are commonly used for biomaterial testing and highlights the possibilities and drawbacks of in vitro tests for biocompatibility evaluation of novel scaffolds. Finally, the review summarizes recent findings concerning the use of adult mesenchymal stem cells for TEP generation and compares the potential of bone marrow- and adipose tissue-derived stem cells in regenerative medicine applications.
Collapse
Affiliation(s)
- Agata Przekora
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland.
| |
Collapse
|
41
|
Osborn J, Aliabouzar M, Zhou X, Rao R, Zhang LG, Sarkar K. Enhanced Osteogenic Differentiation of Human Mesenchymal Stem Cells Using Microbubbles and Low Intensity Pulsed Ultrasound on 3D Printed Scaffolds. ACTA ACUST UNITED AC 2018; 3:e1800257. [PMID: 32627376 DOI: 10.1002/adbi.201800257] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/08/2018] [Indexed: 11/05/2022]
Abstract
Lipid-coated microbubbles, clinically approved as contrast enhancing agents for ultrasound imaging, are investigated for the first time for their possible applications in bone tissue engineering. Effects of microbubbles (average diameter 1.1 µm) coated by a mixture of lipids (1,2-dipalmitoyl-sn-glycero-3-phosphocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000], and 1,2-dipalmitoyl-3-trimethylmmonium-propane) in the presence of low intensity pulsed ultrasound (LIPUS) on human mesenchymal stem cells seeded on 3D printed poly(lactic acid) porous scaffolds are investigated. LIPUS stimulation (30 mW cm-2 , 1.5 MHz, 20% duty cycle) for 3 min a day with 0.5% v/v microbubbles results in a significant increase in proliferation (up to 19.3%) when compared to control after 1, 3, and 5 d. A 3-week osteogenic differentiation study shows a significant increase in total protein content (up to 27.5%), calcium deposition (up to 4.3%), and alkaline phosphatase activity (up to 43.1%) initiated by LIPUS with and without the presence of microbubbles. The microbubbles are found to remain stable during exposure, and their sustained oscillations demonstrably help focus the LIPUS energy toward enhanced cellular response. Integrating LIPUS and microbubbles promises to be a novel and effective strategy for bone tissue engineering and regeneration therapies.
Collapse
Affiliation(s)
- Jenna Osborn
- Mechanical and Aerospace Engineering, George Washington University, Washington, DC, 20052, USA
| | - Mitra Aliabouzar
- Mechanical and Aerospace Engineering, George Washington University, Washington, DC, 20052, USA
| | - Xuan Zhou
- Mechanical and Aerospace Engineering, George Washington University, Washington, DC, 20052, USA
| | - Raj Rao
- Mechanical and Aerospace Engineering, George Washington University, Washington, DC, 20052, USA.,Orthopaedic Surgery, School of Medicine, George Washington University, Washington, DC, 20052, USA
| | - Lijie Grace Zhang
- Mechanical and Aerospace Engineering, George Washington University, Washington, DC, 20052, USA
| | - Kausik Sarkar
- Mechanical and Aerospace Engineering, George Washington University, Washington, DC, 20052, USA
| |
Collapse
|
42
|
Leng X, Shang J, Gao D, Wu J. Low-intensity pulsed ultrasound promotes proliferation and migration of HaCaT keratinocytes through the PI3K/AKT and JNK pathways. ACTA ACUST UNITED AC 2018; 51:e7862. [PMID: 30365726 PMCID: PMC6207286 DOI: 10.1590/1414-431x20187862] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/04/2018] [Indexed: 12/18/2022]
Abstract
Although the effects of low-intensity pulsed ultrasound (LIPUS) on diverse cell types have been fully studied, the functional role of LIPUS in keratinocytes remains poorly understood. This study aimed to investigate the effects of LIPUS on proliferation and migration of HaCaT cells as well as the regulatory mechanisms associated with signaling pathways. Human HaCaT cells were exposed or not to LIPUS, and cell proliferation and migration were measured by BrdU incorporation assay and Transwell assay, respectively. Expression of proteins associated with proliferation and migration was evaluated by western blot analysis. Expression of key kinases in the PI3K/AKT and JNK pathways was also evaluated by western blot analysis. Effects of LIPUS on the PI3K/AKT and JNK pathways, and whether LIPUS affected HaCaT cells via these two pathways were finally explored. When the parameter of LIPUS (number of cycles) was set at 300, cell viability was the highest after LIPUS stimulation. We then found that the percentage of BrdU positive cells was enhanced by LIPUS, along with up-regulation of cyclinD1, CDK6, CDK4, and VEGF. LIPUS promoted migration, as well as up-regulation of MMP-2 and MMP-9. Phosphorylation levels of key kinases in the PI3K/AKT and JNK pathways were increased by LIPUS. Inhibition of either PI3K/AKT pathway or JNK pathway attenuated effects of LIPUS on HaCaT cells, and co-inhibition of these two pathways showed augmented effects. LIPUS promoted proliferation and migration of HaCaT cells through activating the PI3K/AKT and JNK pathways.
Collapse
Affiliation(s)
- Xiaoyan Leng
- Department of Ultrasound, Chengyang People's Hospital, Qingdao, China
| | - Jing Shang
- Health Management Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Danhui Gao
- Department of Ultrasound, Chengyang People's Hospital, Qingdao, China
| | - Jiang Wu
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
43
|
Aliabouzar M, Zhang GL, Sarkar K. Acoustic and mechanical characterization of 3D-printed scaffolds for tissue engineering applications. Biomed Mater 2018; 13:055013. [DOI: 10.1088/1748-605x/aad417] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
44
|
Shuai C, Yang W, Peng S, Gao C, Guo W, Lai Y, Feng P. Physical stimulations and their osteogenesis-inducing mechanisms. Int J Bioprint 2018; 4:138. [PMID: 33102916 PMCID: PMC7581999 DOI: 10.18063/ijb.v4i2.138] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/09/2018] [Indexed: 12/27/2022] Open
Abstract
Physical stimulations such as magnetic, electric and mechanical stimulation could enhance cell activity and promote bone formation in bone repair process via activating signal pathways, modulating ion channels, regulating bonerelated gene expressions, etc. In this paper, bioeffects of physical stimulations on cell activity, tissue growth and bone healing were systematically summarized, which especially focused on their osteogenesis-inducing mechanisms. Detailedly, magnetic stimulation could produce Hall effect which improved the permeability of cell membrane and promoted the migration of ions, especially accelerating the extracellular calcium ions to pass through cell membrane. Electric stimulation could induce inverse piezoelectric effect which generated electric signals, accordingly up-regulating intracellular calcium levels and growth factor synthesis. And mechanical stimulation could produce mechanical signals which were converted into corresponding biochemical signals, thus activating various signaling pathways on cell membrane and inducing a series of gene expressions. Besides, bioeffects of physical stimulations combined with bone scaffolds which fabricated using 3D printing technology on bone cells were discussed. The equipments of physical stimulation system were described. The opportunities and challenges of physical stimulations were also presented from the perspective of bone repair.
Collapse
Affiliation(s)
- Cijun Shuai
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China.,Jiangxi University of Science and Technology, Ganzhou, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Wenjing Yang
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Shuping Peng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Chengde Gao
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Wang Guo
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Yuxiao Lai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Pei Feng
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| |
Collapse
|