1
|
Wen X, Pei F, Jin Y, Zhao Z. Exploring the mechanical and biological interplay in the periodontal ligament. Int J Oral Sci 2025; 17:23. [PMID: 40169537 PMCID: PMC11962160 DOI: 10.1038/s41368-025-00354-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/02/2025] [Accepted: 02/12/2025] [Indexed: 04/03/2025] Open
Abstract
The periodontal ligament (PDL) plays a crucial role in transmitting and dispersing occlusal force, acting as mechanoreceptor for muscle activity during chewing, as well as mediating orthodontic tooth movement. It transforms mechanical stimuli into biological signals, influencing alveolar bone remodeling. Recent research has delved deeper into the biological and mechanical aspects of PDL, emphasizing the importance of understanding its structure and mechanical properties comprehensively. This review focuses on the latest findings concerning both macro- and micro- structural aspects of the PDL, highlighting its mechanical characteristics and factors that influence them. Moreover, it explores the mechanotransduction mechanisms of PDL cells under mechanical forces. Structure-mechanics-mechanotransduction interplay in PDL has been integrated ultimately. By providing an up-to-date overview of our understanding on PDL at various scales, this study lays the foundation for further exploration into PDL-related biomechanics and mechanobiology.
Collapse
Affiliation(s)
- Xinyu Wen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fang Pei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ying Jin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Sun Q, Qiu T, Liu X, Wei Q. Cellular Spatial Sensing Determines Cell Mechanotransduction Activity on the Aligned Nanofibers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410351. [PMID: 39967397 DOI: 10.1002/smll.202410351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/06/2025] [Indexed: 02/20/2025]
Abstract
The geometric properties of extracellular matrix (ECM) fibers play a crucial role in regulating cellular behaviors and functions. Although extensive research has examined the effects of fiber alignment, conflicting results have often arisen, leaving the precise mechanisms by which electrospun fiber alignment affects cellular behavior still unclear. This study investigates how the arrangement of polycaprolactone (PCL) electrospun fiber substrates affects cellular mechanosensing by modulating cell positioning. Larger cells, whose width on a coverslip exceeds 5 times the width of the aligned fiber gaps (≈8 µm in this study) and that span multiple aligned fibers, demonstrate enhanced spreading and mechanotransduction. Conversely, smaller cells, whose width is less than or equal to 2.5 times the width of the aligned fiber gaps and are confined within fiber interstices, exhibit limited mechanotransductive signaling. These findings are further supported by manipulating cell size and, more importantly, have led to the fabrication of semi-aligned fiber networks that enhance both cell spreading and mechanotransduction. This research emphasizes the importance of optimizing fiber architecture to improve cellular interactions, offering valuable insights for the design of biomimetic scaffolds in tissue regeneration.
Collapse
Affiliation(s)
- Qian Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Tiecheng Qiu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Xiaojing Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, and Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, and Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, 250012, P. R. China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, P. R. China
| |
Collapse
|
3
|
Hwangbo H, Chae S, Ryu D, Kim G. In situ magnetic-field-assisted bioprinting process using magnetorheological bioink to obtain engineered muscle constructs. Bioact Mater 2025; 45:417-433. [PMID: 39697238 PMCID: PMC11653149 DOI: 10.1016/j.bioactmat.2024.11.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/17/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Tissue-engineered anisotropic cell constructs are promising candidates for treating volumetric muscle loss (VML). However, achieving successful cell alignment within macroscale 3D cell constructs for skeletal muscle tissue regeneration remains challenging, owing to difficulties in controlling cell arrangement within a low-viscosity hydrogel. Herein, we propose the concept of a magnetorheological bioink to manipulate the cellular arrangement within a low-viscosity hydrogel. This bioink consisted of gelatin methacrylate (GelMA), iron oxide nanoparticles, and human adipose stem cells (hASCs). The cell arrangement is regulated by the responsiveness of iron oxide nanoparticles to external magnetic fields. A bioprinting process using ring magnets was developed for in situ bioprinting, resulting in well-aligned 3D cell structures and enhanced mechanotransduction effects on hASCs. In vitro analyses revealed upregulation of cellular activities, including myogenic-related gene expression, in hASCs. When implanted into a VML mouse model, the bioconstructs improved muscle functionality and regeneration, validating the effectiveness of the proposed approach.
Collapse
Affiliation(s)
- Hanjun Hwangbo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - SooJung Chae
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - GeunHyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do, 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
4
|
Tang K, Tian J, Xu Y, Shang G, Peng X, Yue P, Wang Y, Chen S, Hu Z. Aflatoxin B1 Exposure Suppresses the Migration of Dendritic Cells by Reshaping the Cytoskeleton. Int J Mol Sci 2025; 26:1725. [PMID: 40004187 PMCID: PMC11854954 DOI: 10.3390/ijms26041725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Exposure to Aflatoxin B1 (AFB1) is considered a significant risk factor for human diseases, including the immune function impairment of immune cells. Dendritic cells (DCs), as essential antigen-presenting cells, play a pivotal role in bridging innate and adaptive immunity. However, the impact of AFB1 exposure on DCs has not been fully elucidated. In this study, we investigated the effects of AFB1 exposure on the migration ability of DCs and its underlying action model. Initially, we observed that AFB1 exposure inhibited the survival of DCs and altered their cellular morphology. Further investigation revealed that AFB1 promotes cell adhesion and inhibits DC migration by modulating the expression of cell adhesion molecules. Additionally, our findings indicated that cytoskeletal remodeling plays a crucial role in these processes. Experimental techniques such as immunofluorescence and RNA sequencing confirmed that AFB1 exposure regulates the expression of cytoskeleton-related genes. Moreover, we found that the perturbation of the gene expression profile through AFB1 exposure is associated with cell communication. Collectively, our study findings demonstrate that AFB1 can disrupt the expression of cytoskeleton- and adhesion-related molecules in DCs, thereby altering cell morphology and migration. These insights could provide new perspectives for further understanding the immunosuppressive effects of AFB1 and developing therapeutic strategies for diseases associated with AFB1 exposure.
Collapse
Affiliation(s)
- Kaiyi Tang
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
| | - Jiaxiong Tian
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
| | - Yujun Xu
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
- Key Laboratory of Infectious Immune and Antibody Engineering in University of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Guizhou Medical University, Guiyang 550025, China;
| | - Guofu Shang
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
- Key Laboratory of Infectious Immune and Antibody Engineering in University of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Guizhou Medical University, Guiyang 550025, China;
| | - Xiaoyan Peng
- Key Laboratory of Infectious Immune and Antibody Engineering in University of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Guizhou Medical University, Guiyang 550025, China;
| | - Ping Yue
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
| | - Yun Wang
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
| | - Sen Chen
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
| | - Zuquan Hu
- Immune Cells and Antibody Engineering Research Center in University of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (School of Modern Industry for Health and Medicine)/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; (K.T.); (J.T.); (Y.X.); (G.S.); (P.Y.); (Y.W.)
- Key Laboratory of Infectious Immune and Antibody Engineering in University of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, Guizhou Medical University, Guiyang 550025, China;
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
5
|
Zhou Y, Guo Y, Zhang M, Quan S, Li J. The role of RAP2 in regulation of cell volume on bone marrow mesenchymal stem cell fate determination. J Mol Histol 2025; 56:79. [PMID: 39903386 DOI: 10.1007/s10735-025-10362-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
The extracellular matrix guides cell behavior through mechanical properties, which plays a role in determining cell function and can even influence stem cell fate. Compared with adherent culture, the three-dimensional culture environment is closer to the growth conditions in vivo, but is limited by standardization of material properties and observation and measurement methods. Therefore, it is necessary to study the relationship among the three-dimensional morphological characteristics of cells, cytoskeleton, and stem cell differentiation under adherent culture conditions. Here, we control the cell volume by adjusting the cell density, microfilament cytoskeleton tension, and osmotic pressure of the culture environment, and analyze the cell morphological features and differentiation to the osteoblastic and adipogenic lineages. Based on the in vitro and in vivo results, we identify cell volume as the true reflection of the cytoskeleton tension under stress stimuli compared with cell spreading area. By adjusting cell volume, cytoskeletal tension and cell differentiation can be regulated without affecting cell spreading area. Further study shows that the Ras-related small GTPase RAP2 inhibits the activity of mechanical transducers Lamin A/C and YAP1, playing an important role in cell volume regulation of cell differentiation. In summary, our results support the close relationship between cell volume and cytoskeleton tension. The regulatory role of cell volume on cell differentiation is modulated, at least in part, by RAP2-related mechanosensitive pathways. Our insights into how cell volume regulates cell differentiation may build a bridge between two-dimensional and three-dimensional mechanical studies in cell biology.
Collapse
Affiliation(s)
- Yimei Zhou
- State Key Laboratory of Oral Diseases, National Center of Stomatology, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, 14#, 3rd Section, Renmin South Road, Chengdu, 610041, China
| | - Yutong Guo
- Department of Orthodontics, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, Beijing, 100081, PR China
| | - Mei Zhang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, 14#, 3rd Section, Renmin South Road, Chengdu, 610041, China
| | - Shuqi Quan
- State Key Laboratory of Oral Diseases, National Center of Stomatology, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, 14#, 3rd Section, Renmin South Road, Chengdu, 610041, China
| | - Juan Li
- State Key Laboratory of Oral Diseases, National Center of Stomatology, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, 14#, 3rd Section, Renmin South Road, Chengdu, 610041, China.
| |
Collapse
|
6
|
Fan H, Zhao H, Hou Y, Meng D, Jiang J, Lee EB, Fu Y, Zhang X, Chen R, Wang Y. Heterogeneous focal adhesion cytoskeleton nanoarchitectures from microengineered interfacial curvature to oversee nuclear remodeling and mechanotransduction of mesenchymal stem cells. Cell Mol Biol Lett 2025; 30:10. [PMID: 39856556 PMCID: PMC11762875 DOI: 10.1186/s11658-025-00692-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Interfacial heterogeneity is widely explored to reveal molecular mechanisms of force-mediated pathways due to biased tension. However, the influence of cell density,, curvature, and interfacial heterogeneity on underlying pathways of mechanotransduction is obscure. METHODS Polydimethylsiloxane (PDMS)-based stencils were micropatterned to prepare the micropores for cell culture. The colonies of human mesenchymal stem cells (hMSCs) were formed by controlling cell seeding density to investigate the influences of cell density, curvature and heterogeneity on mechanotransduction. Immunofluorescent staining of integrin, vinculin, and talin-1 was conducted to evaluate adhesion-related expression levels. Then, immunofluorescent staining of actin, actinin, and myosin was performed to detect cytoskeleton distribution, especially at the periphery. Nuclear force-sensing mechanotransduction was explained by yes-associated protein (YAP) and laminA/C analysis. RESULTS The micropatterned colony of hMSCs demonstrated the coincident characters with engineered micropores of microstencils. The cell colony obviously developed the heterogeneous morphogenesis. Heterogeneous focal adhesion guided the development of actin, actinin, and myosin together to regulate cellular contractility and movement by integrin, vinculin, and talin-1. Cytoskeletal staining showed that actin, actinin, and myosin fibers were reorganized at the periphery of microstencils. YAP nuclear translocation and laminA/C nuclear remodeling were enhanced at the periphery by the regulation of heterogeneous focal adhesion (FA) and cytoskeleton arrangement. CONCLUSIONS The characters of the engineered clustering colony showed similar results with prepared microstencils, and colony curvature was also well adjusted to establish heterogeneous balance at the periphery of cell colony. The mechanism of curvature, spreading, and elongation was also investigated to disclose the compliance of FA and cytoskeleton along with curvature microarrays for increased nuclear force-sensing mechanotransduction. The results may provide helpful information for understanding interfacial heterogeneity and nuclear mechanotransduction of stem cells.
Collapse
Affiliation(s)
- Huayu Fan
- Luoyang Orthopedic-Traumatological Hospital Of Henan Province (Henan Provincial Orthopedic Hospital), Zhengzhou, 450008, Henan, China
| | - Hui Zhao
- Zhengzhou Revogene Technology Co., LTD, Airport District, Zhengzhou, 451162, Henan, China
| | - Yan Hou
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Danni Meng
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jizong Jiang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Eon-Bee Lee
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 48513, Republic of Korea
| | - Yinzheng Fu
- Zhengzhou Revogene Technology Co., LTD, Airport District, Zhengzhou, 451162, Henan, China
| | - Xiangdong Zhang
- Luoyang Orthopedic-Traumatological Hospital Of Henan Province (Henan Provincial Orthopedic Hospital), Zhengzhou, 450008, Henan, China.
| | - Rui Chen
- Luoyang Orthopedic-Traumatological Hospital Of Henan Province (Henan Provincial Orthopedic Hospital), Zhengzhou, 450008, Henan, China.
- School of Nursing, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China.
| | - Yongtao Wang
- School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
7
|
Lv JC, Yang X, Zheng ZL, Wang ZG, Hong R, Liu Y, Luo E, Gou JX, Li L, Yuan B, Xu JZ, Li ZM. Engineering Surface-Adaptive Metal-Organic Framework Armor to Promote Infected Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2025; 17:776-789. [PMID: 39689966 DOI: 10.1021/acsami.4c20219] [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: 12/19/2024]
Abstract
Metal-organic frameworks (MOFs) hold enormous promise for treating bacterial infections to circumvent the threat of antibiotic resistance. However, positioning MOFs on wound dressings is hindered and remains a significant challenge. Herein, a facile heterointerfacial engineering strategy was developed to tailor the "MOF armor" that adaptively weaponized the poly(ε-caprolactone) electrospun dressing with excellent bacteria-killing efficacy. Hydrophilic epitaxial crystallization to enhance the interfacial wettability is the key to induce the uniform seeding of Cu2+ and thus to generate a compact MOF layer on the electrospun dressing. The universality of the proposed strategy was demonstrated by the construction of different kinds of MOFs (HKUST-1, ZIF-8, and ZIF-67) on variously shaped substrates (nanofibers, pellets, plates, and 3D-printed porous scaffolds). By optimizing the Cu2+ loading, the Cu-MOF armor exhibited sustained ion release behavior, strong antibacterial activity, and good biocompatibility. In vivo rat model revealed that the Cu-MOF armor significantly promoted infected wound healing by inhibiting inflammatory factors, promoting collagen deposition, and angiogenesis. This unique MOF armor provides an appealing and effective solution for designing and fabricating advanced wound dressings.
Collapse
Affiliation(s)
- Jia-Cheng Lv
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Xue Yang
- West China Hospital, Sichuan University West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Zi-Li Zheng
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zhi-Guo Wang
- West China Hospital, Sichuan University West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Rui Hong
- West China Hospital, Sichuan University West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ju-Xiang Gou
- Thyroid Surgery Department, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lingli Li
- West China Hospital, Sichuan University West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Bo Yuan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
- West China Hospital, Sichuan University West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Zhong-Ming Li
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
- West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
He S, Zhang Q, Jia J, Xia W, Chen S, Min F, Song Y, Yu Y, Li J, Li Z, Luo G. Stiffness and surface topology of silicone implants competitively mediate inflammatory responses of macrophages and foreign body response. Mater Today Bio 2024; 29:101304. [PMID: 39498150 PMCID: PMC11532915 DOI: 10.1016/j.mtbio.2024.101304] [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: 06/07/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Adverse inflammatory responses, dominated by macrophages, that are induced by physical cues of silicone implants can heavily damage the life quality of patients via causing fibrosis and device failure. As stiffness and surface topology affect macrophages at the same time, the competition or partnership among physical cues against the regulation of macrophages is still ambiguous. Herein, a series of PDMS implants with different stiffness at ∼ MPa and surface topology at tens of micrometers were fabricated to investigate the relationship, the regulation rule, and the underlying mechanism of the two physical cues against the inflammatory responses of M1 macrophages. There is a competitive rule: surface topology could suppress the inflammatory responses of M1 macrophages in the soft group but did not have the same effect in the stiff group. Without surface topology, lower stiffness unexpectedly evoked stronger inflammatory responses of M1 macrophages. Implanting experiments also proved that the competitive state against mediating in vivo immune responses and the unexpected inflammatory responses. The reason is that stiffness could strongly up-regulate focal adhesion and activate the MAPK/NF-κB signaling axis to evoke inflammatory responses, which could shield the effect of surface topology. Therefore, for patient healthcare, it is crucial to prioritize stiffness while not surface topology at MPa levels to minimize adverse reactions.
Collapse
Affiliation(s)
- Sicen He
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Qingrong Zhang
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jiezhi Jia
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Wei Xia
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Shengnan Chen
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing, 100190, PR China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Fanyi Min
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing, 100190, PR China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yanlin Song
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing, 100190, PR China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yunlong Yu
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jiangfeng Li
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Zheng Li
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Gaoxing Luo
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| |
Collapse
|
9
|
Fang L, Lin X, Xu R, Liu L, Zhang Y, Tian F, Li JJ, Xue J. Advances in the Development of Gradient Scaffolds Made of Nano-Micromaterials for Musculoskeletal Tissue Regeneration. NANO-MICRO LETTERS 2024; 17:75. [PMID: 39601962 PMCID: PMC11602939 DOI: 10.1007/s40820-024-01581-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
The intricate hierarchical structure of musculoskeletal tissues, including bone and interface tissues, necessitates the use of complex scaffold designs and material structures to serve as tissue-engineered substitutes. This has led to growing interest in the development of gradient bone scaffolds with hierarchical structures mimicking the extracellular matrix of native tissues to achieve improved therapeutic outcomes. Building on the anatomical characteristics of bone and interfacial tissues, this review provides a summary of current strategies used to design and fabricate biomimetic gradient scaffolds for repairing musculoskeletal tissues, specifically focusing on methods used to construct compositional and structural gradients within the scaffolds. The latest applications of gradient scaffolds for the regeneration of bone, osteochondral, and tendon-to-bone interfaces are presented. Furthermore, the current progress of testing gradient scaffolds in physiologically relevant animal models of skeletal repair is discussed, as well as the challenges and prospects of moving these scaffolds into clinical application for treating musculoskeletal injuries.
Collapse
Affiliation(s)
- Lei Fang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Xiaoqi Lin
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Ruian Xu
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Lu Liu
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yu Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Feng Tian
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| | - Jiajia Xue
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| |
Collapse
|
10
|
Jiang K, Luo C, Li YM, Wang K, Huang S, You XH, Liu Y, Luo E, Xu JZ, Zhang L, Li ZM. An immunomodulatory and osteogenic bacterial cellulose scaffold for bone regeneration via regulating the immune microenvironment. Int J Biol Macromol 2024; 281:136375. [PMID: 39383912 DOI: 10.1016/j.ijbiomac.2024.136375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Creating a bone homeostasis microenvironment that balances osteogenesis and immunity is a substantial challenge for bone regeneration. Here, we prepared an immunomodulatory and osteogenic bacterial cellulose scaffold (FOBS) via a facile one-pot approach. The aldehyde groups were generated via selective oxidation of the hydroxyl groups of bacterial cellulose, offering the bonding sites for dopamine through a Schiff base reaction. At the same time, the deposition of Ca2+ and PO43- was promoted on the aldehyde cellulose scaffold because of the high affinity of the catechol moiety for Ca2+. Compared with that of the unmodified scaffold, the hydroxyapatite content of FOBS increased by 47.1 % according to the ICP results. Interestingly, FOBS regulated the immune microenvironment to accelerate the conversion of M1 to M2 macrophages. The expressions of ARG-1 and Dectin-1 (M2) in the FOBS group increased by >100 %. The expression of osteogenic differentiation of BMSCs was also upregulated. In a rat cranial defect model, the BV/TV of FOBS was significantly increased. Further immunohistochemical analysis revealed that an improved immune microenvironment promoted the osteogenic differentiation of stem cells in vivo. This work provides an effective and easy-to-operate strategy for the development of the bone tissue engineering scaffolds.
Collapse
Affiliation(s)
- Kai Jiang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Chuan Luo
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yuan-Min Li
- Key Laboratory of Transplant Engineering and Immunology, NHC, Chengdu 610065, China; Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Kai Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Shishu Huang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Xuan-He You
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Li Zhang
- Department of Rehabilitation Medicine, West China Second Hospital, Sichuan University, Chengdu 610065, China.
| | - Zhong-Ming Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
11
|
Liu K, Li L, Li Y, Luo Y, Zhang Z, Wen W, Ding S, Huang Y, Liu M, Zhou C, Luo B. Creating a bionic scaffold via light-curing liquid crystal ink to reveal the role of osteoid-like microenvironment in osteogenesis. Bioact Mater 2024; 40:244-260. [PMID: 38973990 PMCID: PMC11226751 DOI: 10.1016/j.bioactmat.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 07/09/2024] Open
Abstract
Osteoid plays a crucial role in directing cell behavior and osteogenesis through its unique characteristics, including viscoelasticity and liquid crystal (LC) state. Thus, integrating osteoid-like features into 3D printing scaffolds proves to be a promising approach for personalized bone repair. Despite extensive research on viscoelasticity, the role of LC state in bone repair has been largely overlooked due to the scarcity of suitable LC materials. Moreover, the intricate interplay between LC state and viscoelasticity in osteogenesis remains poorly understood. Here, we developed innovative hydrogel scaffolds with osteoid-like LC state and viscoelasticity using digital light processing with a custom LC ink. By utilizing these LC scaffolds as 3D research models, we discovered that LC state mediates high protein clustering to expose accessible RGD motifs to trigger cell-protein interactions and osteogenic differentiation, while viscoelasticity operates via mechanotransduction pathways. Additionally, our investigation revealed a synergistic effect between LC state and viscoelasticity, amplifying cell-protein interactions and osteogenic mechanotransduction processes. Furthermore, the interesting mechanochromic response observed in the LC hydrogel scaffolds suggests their potential application in mechanosensing. Our findings shed light on the mechanisms and synergistic effects of LC state and viscoelasticity in osteoid on osteogenesis, offering valuable insights for the biomimetic design of bone repair scaffolds.
Collapse
Affiliation(s)
- Kun Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, PR China
| | - Lin Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, PR China
| | - Yizhi Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, PR China
| | - Yiting Luo
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, PR China
| | - Zhaoyu Zhang
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, PR China
| | - Wei Wen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, PR China
| | - Shan Ding
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, PR China
| | - Yadong Huang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Guangzhou, 510632, PR China
| | - Mingxian Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, PR China
| | - Changren Zhou
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, PR China
| | - Binghong Luo
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, PR China
| |
Collapse
|
12
|
Wang H, Weng X, Chen Y, Mao S, Gao Y, Wu Q, Huang Y, Guan X, Xu Z, Lai Y. Biomimetic concentric microgrooved titanium surfaces influence bone marrow-derived mesenchymal stem cell osteogenic differentiation via H3K4 trimethylation epigenetic regulation. Dent Mater J 2024; 43:683-692. [PMID: 39135261 DOI: 10.4012/dmj.2023-327] [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: 10/01/2024]
Abstract
Material surface micromorphology can modulate cellular behavior and promote osteogenic differentiation through cytoskeletal rearrangement. Bone reconstruction requires precise regulation of gene expression in cells, a process governed by epigenetic mechanisms such as histone modifications, DNA methylation, and chromatin remodeling. We constructed osteon-mimetic concentric microgrooved titanium surfaces with different groove sizes and cultured bone marrow-derived mesenchymal stem cells (BMSCs) on the material surfaces to study how they regulate cell biological behavior and osteogenic differentiation through epigenetics. We found that the cells arranged in concentric circles along the concentric structure in the experimental group, and the concentric microgrooved surface did not inhibit cell proliferation. The results of a series of osteogenic differentiation experiments showed that the concentric microgrooves facilitated calcium deposition and promoted osteogenic differentiation of the BMSCs. Concentric microgrooved titanium surfaces that were 30 μm wide and 10 μm deep promoted osteogenic differentiation of BMSC by increasing WDR5 expression via H3K4 trimethylation upregulation.
Collapse
Affiliation(s)
- Hong Wang
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
- Stomatological Hospital of Xiamen Medical college
| | - Xinze Weng
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Yan Chen
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Shunjie Mao
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Yuerong Gao
- Department of Stomatology of The Third Affiliated Hospital of Xi'an Medical University
| | - Qinglin Wu
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Yanling Huang
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Xin Guan
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Zhiqiang Xu
- Department of Stomatology, Affiliated Hospital of Putian University
| | - Yingzhen Lai
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| |
Collapse
|
13
|
Chen YQ, Lee HC, Lee HH. Shp2 contributes to the regulation of nuclear shape and cellular viscoelasticity in response to substrate spatial cues. Biochem Biophys Res Commun 2024; 721:150144. [PMID: 38781661 DOI: 10.1016/j.bbrc.2024.150144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 04/19/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Cell polarization can be guided by substrate topology through space constraints and adhesion induction, which are part of cellular mechanosensing pathways. Here, we demonstrated that protein tyrosine phosphatase Shp2 plays a crucial role in mediating the response of cells to substrate spatial cues. When compared to cells spreading on surfaces coated uniformly with fibronectin (FN), cells attached to 10 μm-width FN-strip micropattern (MP), which provides spatial cues for uniaxial spreading, exhibited elongated focal adhesions (FAs) and aligned stress fibers in the direction of the MP. As a result of uniaxial cell spreading, nuclei became elongated, dependent on ROCK-mediated actomyosin contractility. Additionally, intracellular viscoelasticity also increased. Shp2-deficient cells did not display elongated FAs mediated by MP, well-aligned stress fibers, or changes in nuclear shape and intracellular viscoelasticity. Overall, our data suggest that Shp2 is involved in regulating FAs and the actin cytoskeleton to modulate nuclear shape and intracellular physical properties in response to substrate spatial cues.
Collapse
Affiliation(s)
- Yin-Quan Chen
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsin-Chang Lee
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Hui Lee
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
14
|
Wei X, Chen J, Shen HY, Jiang K, Ren H, Liu Y, Luo E, Zhang J, Xu JZ, Li ZM. Hierarchically Biomimetic Scaffolds with Anisotropic Micropores and Nanotopological Patterns to Promote Bone Regeneration via Geometric Modulation. Adv Healthc Mater 2024; 13:e2304178. [PMID: 38490686 DOI: 10.1002/adhm.202304178] [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: 11/27/2023] [Revised: 02/29/2024] [Indexed: 03/17/2024]
Abstract
Structural engineering is an appealing means to modulate osteogenesis without the intervention of exogenous cells or therapeutic agents. In this work, a novel 3D scaffold with anisotropic micropores and nanotopographical patterns is developed. Scaffolds with oriented pores are fabricated via the selective extraction of water-soluble polyethylene oxide from its poly(ε-caprolactone) co-continuous mixture and uniaxial stretching. The plate apatite-like lamellae are subsequently hatched on the pore walls through surface-induced epitaxial crystallization. Such a unique geometric architecture yields a synergistic effect on the osteogenic capability. The prepared scaffold leads to a 19.2% and 128.0% increase in the alkaline phosphatase activity of rat bone mesenchymal stem cells compared to that of the scaffolds with only oriented pores and only nanotopographical patterns, respectively. It also induces the greatest upregulation of osteogenic-related gene expression in vitro. The cranial defect repair results demonstrate that the prepared scaffold effectively promotes new bone regeneration, as indicated by a 350% increase in collagen I expression in vivo compared to the isotropic porous scaffold without surface nanotopology after implantation for 14 weeks. Overall, this work provides geometric motifs for the transduction of biophysical cues in 3D porous scaffolds, which is a promising option for tissue engineering applications.
Collapse
Affiliation(s)
- Xin Wei
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Jiaxin Chen
- Center for Plastic & Reconstructive Surgery, Department of Plastic & Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, P. R. China
| | - Hui-Yuan Shen
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Kai Jiang
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Haohao Ren
- College of Physics, Sichuan University, Chengdu, 610064, P. R. China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Jin Zhang
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, P. R. China
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, P. R. China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, P. R. China
| | - Zhong-Ming Li
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, P. R. China
| |
Collapse
|
15
|
Jiang K, Wang K, Luo C, Su BY, Du H, Liu Y, Lei J, Luo E, Cardon L, Edeleva M, Huang SS, Xu JZ, Li ZM. A Biomimetic Fibrous Composite Scaffold with Nanotopography-Regulated Mineralization for Bone Defect Repair. Biomacromolecules 2024; 25:3784-3794. [PMID: 38743836 DOI: 10.1021/acs.biomac.4c00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The effective regeneration of large bone defects via bone tissue engineering is challenging due to the difficulty in creating an osteogenic microenvironment. Inspired by the fibrillar architecture of the natural extracellular matrix, we developed a nanoscale bioengineering strategy to produce bone fibril-like composite scaffolds with enhanced osteogenic capability. To activate the surface for biofunctionalization, self-adaptive ridge-like nanolamellae were constructed on poly(ε-caprolactone) (PCL) electrospinning scaffolds via surface-directed epitaxial crystallization. This unique nanotopography with a markedly increased specific surface area offered abundant nucleation sites for Ca2+ recruitment, leading to a 5-fold greater deposition weight of hydroxyapatite than that of the pristine PCL scaffold under stimulated physiological conditions. Bone marrow mesenchymal stem cells (BMSCs) cultured on bone fibril-like scaffolds exhibited enhanced adhesion, proliferation, and osteogenic differentiation in vitro. In a rat calvarial defect model, the bone fibril-like scaffold significantly accelerated bone regeneration, as evidenced by micro-CT, histological histological and immunofluorescence staining. This work provides the way for recapitulating the osteogenic microenvironment in tissue-engineered scaffolds for bone repair.
Collapse
Affiliation(s)
- Kai Jiang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Kai Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Chuan Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Biao-Yao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Hao Du
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - Jun Lei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - Ludwig Cardon
- Centre for Polymer and Material Technologies, Department of Materials Textiles and Chemical Engineering, Ghent University, Technologiepark-Zwijnaarde 130, Gent 9052, Belgium
| | - Mariya Edeleva
- Centre for Polymer and Material Technologies, Department of Materials Textiles and Chemical Engineering, Ghent University, Technologiepark-Zwijnaarde 130, Gent 9052, Belgium
| | - Shi-Shu Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zhong-Ming Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
16
|
Lai H, Huo X, Han Y, Hu M, Kong X, Yin J, Qian J. Electrowriting patterns and electric field harness directional cell migration for skin wound healing. Mater Today Bio 2024; 26:101083. [PMID: 38757058 PMCID: PMC11097089 DOI: 10.1016/j.mtbio.2024.101083] [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: 01/25/2024] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
Directional cell migration is a crucial step in wound healing, influenced by electrical and topographic stimulations. However, the underlying mechanism and the combined effects of these two factors on cell migration remain unclear. This study explores cell migration under various combinations of guided straight line (SL) spacing, conductivity, and the relative direction of electric field (EF) and SL. Electrowriting is employed to fabricate conductive (multiwalled carbon nanotube/polycaprolactone (PCL)) and nonconductive (PCL) SL, with narrow (50 μm) and wide (400 μm) spacing that controls the topographic stimulation strength. Results show that various combinations of electrical and topographic stimulation yield significantly distinct effects on cell migration direction and speed; cells migrate fastest with the most directivity in the case of conductive, narrow-spacing SL parallel to EF. A physical model based on intercellular interactions is developed to capture the underlying mechanism of cell migration under SL and EF stimulations, in agreement with experimental observations. In vivo skin wound healing assay further confirmed that the combination of EF (1 V cm-1) and parallelly aligned conductive fibers accelerated the wound healing process. This study presents a promising approach to direct cell migration and enhance wound healing by optimizing synergistic electrical and topographic stimulations.
Collapse
Affiliation(s)
- Huinan Lai
- Department of Engineering Mechanics, Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Xiaodan Huo
- The State Key Laboratory of Fluid Power Transmission and Control Systems, Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
| | - Ying Han
- The State Key Laboratory of Fluid Power Transmission and Control Systems, Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
| | - Minyu Hu
- Department of Engineering Mechanics, Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Xiangren Kong
- Department of Engineering Mechanics, Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Jun Yin
- The State Key Laboratory of Fluid Power Transmission and Control Systems, Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
| | - Jin Qian
- Department of Engineering Mechanics, Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Zhejiang University, Hangzhou, China
- The State Key Laboratory of Fluid Power Transmission and Control Systems, Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Li Y, Tang S, Luo Z, Liu K, Luo Y, Wen W, Ding S, Li L, Liu M, Zhou C, Luo B. Chitin whisker/chitosan liquid crystal hydrogel assisted scaffolds with bone-like ECM microenvironment for bone regeneration. Carbohydr Polym 2024; 332:121927. [PMID: 38431420 DOI: 10.1016/j.carbpol.2024.121927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/03/2024] [Accepted: 02/06/2024] [Indexed: 03/05/2024]
Abstract
Natural bone exhibits a complex anisotropic and micro-nano hierarchical structure, more importantly, bone extracellular matrix (ECM) presents liquid crystal (LC) phase and viscoelastic characteristics, providing a unique microenvironment for guiding cell behavior and regulating osteogenesis. However, in bone tissue engineering scaffolds, the construction of bone-like ECM microenvironment with exquisite microstructure is still a great challenge. Here, we developed a novel polysaccharide LC hydrogel supported 3D printed poly(l-lactide) (PLLA) scaffold with bone-like ECM microenvironment and micro-nano aligned structure. First, we prepared a chitin whisker/chitosan polysaccharide LC precursor, and then infuse it into the pores of 3D printed PLLA scaffold, which was previously surface modified with a polydopamine layer. Next, the LC precursor was chemical cross-linked by genipin to form a hydrogel network with bone-like ECM viscoelasticity and LC phase in the scaffold. Subsequently, we performed directional freeze-casting on the composite scaffold to create oriented channels in the LC hydrogel. Finally, we soaked the composite scaffold in phytic acid to further physical cross-link the LC hydrogel through electrostatic interactions and impart antibacterial effects to the scaffold. The resultant biomimetic scaffold displays osteogenic activity, vascularization ability and antibacterial effect, and is expected to be a promising candidate for bone repair.
Collapse
Affiliation(s)
- Yizhi Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Shengyue Tang
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Ziang Luo
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Kun Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Yiting Luo
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Wei Wen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Shan Ding
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Lihua Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Mingxian Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Changren Zhou
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Binghong Luo
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China.
| |
Collapse
|
18
|
Shen HY, Xing F, Shang SY, Jiang K, Kuzmanović M, Huang FW, Liu Y, Luo E, Edeleva M, Cardon L, Huang S, Xiang Z, Xu JZ, Li ZM. Biomimetic Mineralized 3D-Printed Polycaprolactone Scaffold Induced by Self-Adaptive Nanotopology to Accelerate Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:18658-18670. [PMID: 38587811 DOI: 10.1021/acsami.4c02636] [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: 04/09/2024]
Abstract
Three-dimensional (3D)-printed biodegradable polymer scaffolds are at the forefront of personalized constructs for bone tissue engineering. However, it remains challenging to create a biological microenvironment for bone growth. Herein, we developed a novel yet feasible approach to facilitate biomimetic mineralization via self-adaptive nanotopography, which overcomes difficulties in the surface biofunctionalization of 3D-printed polycaprolactone (PCL) scaffolds. The building blocks of self-adaptive nanotopography were PCL lamellae that formed on the 3D-printed PCL scaffold via surface-directed epitaxial crystallization and acted as a linker to nucleate and generate hydroxyapatite crystals. Accordingly, a uniform and robust mineralized layer was immobilized throughout the scaffolds, which strongly bound to the strands and had no effect on the mechanical properties of the scaffolds. In vitro cell culture experiments revealed that the resulting scaffold was biocompatible and enhanced the proliferation and osteogenic differentiation of mouse embryolous osteoblast cells. Furthermore, we demonstrated that the resulting scaffold showed a strong capability to accelerate in vivo bone regeneration using a rabbit bone defect model. This study provides valuable opportunities to enhance the application of 3D-printed scaffolds in bone repair, paving the way for translation to other orthopedic implants.
Collapse
Affiliation(s)
- Hui-Yuan Shen
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Fei Xing
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Si-Yuan Shang
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Kai Jiang
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Maja Kuzmanović
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Fu-Wen Huang
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mariya Edeleva
- Centre for Polymer and Material Technologies, Department of Materials, Textiles and Chemical Engineering, Ghent University, Ghent 9052, Belgium
| | - Ludwig Cardon
- Centre for Polymer and Material Technologies, Department of Materials, Textiles and Chemical Engineering, Ghent University, Ghent 9052, Belgium
| | - Shishu Huang
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhou Xiang
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zhong-Ming Li
- College of Polymer Science and Engineering and State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
19
|
张 欢, 李 卓, 林 敏. [Integrin and N-cadherin Co-Regulate the Polarity of Mesenchymal Stem Cells via Mechanobiological Mechanisms]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:321-329. [PMID: 38645863 PMCID: PMC11026872 DOI: 10.12182/20240360104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Indexed: 04/23/2024]
Abstract
Objective To investigate the synergistic regulation of the polarization of mesenchymal stem cells by integrin and N-cadherin-mediated mechanical adhesion and the underlying mechanobiological mechanisms. Methods Bilayer polyethylene glyeol (PEG) hydrogels were formulated and modified with RGD and HAVDI peptides, respectively, to achieve mechanical adhesion to integrin and N-cadherin and to replicate the integrin-mediated mechanical interaction between cells and the extracellular matrix and the N-cadherin-mediated cell-cell mechanical interaction. The polar proteins, phosphatidylinositol 3-kinase (PI3K) and phosphorylated myosin light chain (pMLC), were characterized through immunofluorescence staining in individual cells with or without contact with HAVDI peptides under integrin-mediated adhesion, N-cadherin-mediated adhesion, and different intracellular forces. Their expression levels and polar distribution were analyzed using Image J. Results Integrin-mediated adhesion induced significantly higher polar strengths of PI3K and pMLC in the contact group than in those in the no contact group, resulting in the concentration of the polar angle of PI3K to β-catenin in the range of 135° to 180° and the concentration of the polar angle of pMLC to β-catenin in the range of 0° to 45° in the contact group. Inhibition of integrin function led to inhibition of the polarity distribution of PI3K in the contact group, but did not change the polarity distribution of pMLC protein. The effect of N-cadherin on the polarity distributions of PI3K and pMLC was similar to that of integrin. However, inhibition of the mechanical adhesion of N-cadherin led to inhibition of the polarity intensity and polarity angle distribution of PI3K and pMLC proteins in the contact group. Furthermore, inhibition of the mechanical adhesion of N-cadherin caused weakened polarity intensity of integrin β1, reducing the proportion of cells with polarity angles between integrin β1 and β-catenin concentrating in the range of 135° to 180°. Additionally, intracellular forces influenced the polar distribution of PI3K and pMLC proteins. Reducing intracellular forces weakened the polarity intensity of PI3K and pMLC proteins and their polarity distribution, while increasing intracellular forces enhanced the polarity intensity of PI3K and pMLC proteins and their polarity distribution. Conclusion Integrin and N-cadherin co-regulate the polarity distribution of cell proteins and N-cadherin can play an important role in the polarity regulation of stem cells through local inhibition of integrin.
Collapse
Affiliation(s)
- 欢 张
- 西安交通大学生命科学与技术学院 生物信息工程教育部重点实验室 (西安 710049)The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- 西安交通大学生命科学与技术学院 仿生工程与生物力学研究所 (西安 710049)Bioinspired Engineering and Biomechanics Center (BEBC), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - 卓雅 李
- 西安交通大学生命科学与技术学院 生物信息工程教育部重点实验室 (西安 710049)The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- 西安交通大学生命科学与技术学院 仿生工程与生物力学研究所 (西安 710049)Bioinspired Engineering and Biomechanics Center (BEBC), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - 敏 林
- 西安交通大学生命科学与技术学院 生物信息工程教育部重点实验室 (西安 710049)The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- 西安交通大学生命科学与技术学院 仿生工程与生物力学研究所 (西安 710049)Bioinspired Engineering and Biomechanics Center (BEBC), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
20
|
Dong X, Su S, Sun Q, Wang P, Hu Q, Wei Q. Aligned Nanofibers Promote Myoblast Polarization and Myogenesis through Activating Rac-Related Signaling Pathways. ACS Biomater Sci Eng 2024; 10:1712-1721. [PMID: 38422457 DOI: 10.1021/acsbiomaterials.4c00127] [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: 03/02/2024]
Abstract
The extracellular matrix (ECM) plays a crucial role in regulating cellular behaviors and functions. However, the impact of ECM topography on muscle cell adhesion and differentiation remains poorly understood from a mechanosensing perspective. In this study, we fabricated aligned and random electrospun polycaprolactone (PCL) nanofibers to mimic the structural characteristics of ECM. Mechanism investigations revealed that the orientation of nanofibers promoted C2C12 polarization and myogenesis through Rac-related signaling pathways. Conversely, cells cultured on random fibers exhibited spreading behavior mediated by RhoA/ROCK pathways, resulting in enhanced stress fiber formation but reduced capacity for myogenic differentiation. Our findings highlight the critical role of an ECM structure in muscle regeneration and damage repair, providing novel insights into mechanosensing mechanisms underlying muscle injury diseases.
Collapse
Affiliation(s)
- Xiangyu Dong
- Department of Gynecology and Obstetrics, Frontiers Science Center for Disease-related Molecular Network, West China Second University Hospital, Sichuan University, Chengdu 610041, P. R. China
- Department of Nephrology, Kidney Research Institute, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Shan Su
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Qian Sun
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Peng Wang
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Qian Hu
- Department of Gynecology and Obstetrics, Frontiers Science Center for Disease-related Molecular Network, West China Second University Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Qiang Wei
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| |
Collapse
|
21
|
Shi N, Wang J, Tang S, Zhang H, Wei Z, Li A, Ma Y, Xu F. Matrix Nonlinear Viscoelasticity Regulates Skeletal Myogenesis through MRTF Nuclear Localization and Nuclear Mechanotransduction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305218. [PMID: 37847903 DOI: 10.1002/smll.202305218] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/30/2023] [Indexed: 10/19/2023]
Abstract
Mechanically sensitive tissues (e.g., skeletal muscles) greatly need mechanical stimuli during the development and maturation. The extracellular matrix (ECM) mediates these signals through nonlinear viscoelasticity of collagen networks that are predominant components of the ECM. However, the interactions between cells and ECM form a feedback loop, and it has not yet been possible to determine the degree to which, if any, of the features of matrix nonlinear viscoelasticity affect skeletal muscle development and regeneration. In this study, a nonlinear viscoelastic feature (i.e., strain-enhanced stress relaxation (SESR)) in normal skeletal muscles is observed, which however is almost absent in diseased muscles from Duchenne muscular dystrophy mice. It is recapitulated such SESR feature in vitro and separated the effects of mechanical strain and ECM viscoelasticity on myoblast response by developing a collagen-based hydrogel platform. Both strain and stress relaxation induce myogenic differentiation and myotube formation by C2C12 myoblasts, and myogenesis is more promoted by applying SESR. This promotion can be explained by the effects of SESR on actin polymerization-mediated myocardin related transcription factor (MRTF) nuclear localization and nuclear mechanotransduction. This study represents the first attempt to investigate the SESR phenomenon in skeletal muscles and reveal underlying mechanobiology, which will provide new opportunities for the tissue injury treatments.
Collapse
Affiliation(s)
- Nianyuan Shi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Jing Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Shaoxin Tang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Hui Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Zhao Wei
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Yufei Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
22
|
Jiao Y, Yang X, Li Y, Wang F, Wang L, Li C. Spider-Silk-like Fiber Mat-Covered Polypropylene Warp-Knitted Hernia Mesh for Inhibition of Fibrosis under Dynamic Environment. Biomacromolecules 2024; 25:1214-1227. [PMID: 38295271 DOI: 10.1021/acs.biomac.3c01181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Hernia surgery is a widely performed procedure, and the use of a polypropylene mesh is considered the standard approach. However, the mesh often leads to complications, including the development of scar tissue that wraps around the mesh and causes it to shrink. Consequently, there is a need to investigate the relationship between the mesh and scar formation as well as to develop a hernia mesh that can prevent fibrosis. In this study, three different commercial polypropylene hernia meshes were examined to explore the connection between the fabric structure and mechanical properties. In vitro dynamic culture was used to investigate the mechanism by which the mechanical properties of the mesh in a dynamic environment affect cell differentiation. Additionally, electrospinning was employed to create polycaprolactone spider-silk-like fiber mats to achieve mechanical energy dissipation in dynamic conditions. These fiber mats were then combined with the preferred hernia mesh. The results demonstrated that the composite mesh could reduce the activation of fibroblast mechanical signaling pathways and inhibit its differentiation into myofibroblasts in dynamic environments.
Collapse
Affiliation(s)
- Yongjie Jiao
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - Xiaowei Yang
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - Yan Li
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - Fujun Wang
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - Lu Wang
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - Chaojing Li
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| |
Collapse
|
23
|
Cong M, Wu X, Zhu L, Gu G, Ding F, Li G, Shi H. Anisotropic microtopography surface of chitosan scaffold regulating skin precursor-derived Schwann cells towards repair phenotype promotes neural regeneration. Regen Biomater 2024; 11:rbae005. [PMID: 38414797 PMCID: PMC10898340 DOI: 10.1093/rb/rbae005] [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: 09/25/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 02/29/2024] Open
Abstract
For repairing peripheral nerve and spinal cord defects, biomaterial scaffold-based cell-therapy was emerged as an effective strategy, requiring the positive response of seed cells to biomaterial substrate and environment signals. Previous work highlighted that the imposed surface properties of scaffold could provide important guidance cues to adhered cells for polarization. However, the insufficiency of native Schwann cells and unclear cellular response mechanisms remained to be addressed. Given that, this study aimed to illuminate the micropatterned chitosan-film action on the rat skin precursor-derived Schwann cells (SKP-SCs). Chitosan-film with different ridge/groove size was fabricated and applied for the SKP-SCs induction. Results indicated that SKP-SCs cultured on 30 μm size microgroove surface showed better oriented alignment phenotype. Induced SKP-SCs presented similar genic phenotype as repair Schwann cells, increasing expression of c-Jun, neural cell adhesion molecule, and neurotrophic receptor p75. Moreover, SKP-SC-secretome was subjected to cytokine array GS67 assay, data indicated the regulation of paracrine phenotype, a panel of cytokines was verified up-regulated at secreted level and gene expression level in induced SKP-SCs. These up-regulated cytokines exhibit a series of promotive neural regeneration functions, including cell survival, cell migration, cell proliferation, angiogenesis, axon growth, and cellular organization etc. through bioinformatics analysis. Furthermore, the effectively polarized SKP-SCs-sourced secretome, promoted the proliferation and migration capacity of the primarily cultured native rat Schwann cells, and augmented neurites growth of the cultured motoneurons, as well as boosted axonal regrowth of the axotomy-injured motoneurons. Taken together, SKP-SCs obtained pro-neuroregeneration phenotype in adaptive response to the anisotropic topography surface of chitosan-film, displayed the oriented parallel growth, the transition towards repair Schwann cell genic phenotype, and the enhanced paracrine effect on neural regeneration. This study provided novel insights into the potency of anisotropic microtopography surface to Schwann-like cells phenotype regulation, that facilitating to provide promising engineered cell-scaffold in neural injury therapies.
Collapse
Affiliation(s)
- Meng Cong
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Xia Wu
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Lingjie Zhu
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong 226001, China
| | - Guohao Gu
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Fei Ding
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Guicai Li
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Haiyan Shi
- Key Laboratory of Neuroregenration of Jiangsu and Ministry of Education and Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong 226001, China
| |
Collapse
|
24
|
江 文, 钟 健, 欧阳 智, 沈 钧, 邱 艳, 曾 烨. [Spatial Constraints of Rectangular Hydrogel Microgrooves Regulate the Morphology and Arrangement of Human Umbilical Vein Endothelial Cells]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:87-94. [PMID: 38322512 PMCID: PMC10839481 DOI: 10.12182/20240160402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Indexed: 02/08/2024]
Abstract
Objective To construct microscale rectangular hydrogel grooves and to investigate the morphology and alignment of human umbilical vein endothelial cells (HUVECs) under spatial constraints. Vascular endothelial cell morphology and alignment are important factors in vascular development and the maintenance of homeostasis. Methods A 4-arm polyethylene glycol-acrylate (PEG-acrylate) hydrogel was used to fabricate rectangular microgrooves of the widths of 60 μm, 100 μm, and 140 μm. The sizes and the fibronectin (FN) adhesion of these hydrogel microgrooves were measured. HUVECs were seeded onto the FN-coated microgrooves, while the flat surface without micropatterns was used as the control. After 48 hours of incubation, the morphology and orientation of the cells were examined. The cytoskeleton was labelled with phalloidine and the orientation of the cytoskeleton in the hydrogel microgrooves was observed by laser confocal microscopy. Results The hydrogel microgrooves constructed exhibited uniform and well-defined morphology, a complete structure, and clear edges, with the width deviation being less than 3.5%. The depth differences between the hydrogel microgrooves of different widths were small and the FN adhesion is uniform, providing a micro-patterned growth interface for cells. In the control group, the cells were arranged haphazardly in random orientations and the cell orientation angle was (46.9±1.8)°. In contrast, the cell orientation angle in the hydrogel microgrooves was significantly reduced (P<0.001). However, the cell orientation angles increased with the increase in hydrogel microgroove width. For the 60 μm, 100 μm, and 140 μm hydrogel microgrooves, the cell orientation angles were (16.4±2.8)°, (24.5±3.2)°, and (30.3±3.5)°, respectively. Compared to that of the control group (35.7%), the number of cells with orientation angles <30° increased significantly in the hydrogel microgrooves of different widths (P<0.001). However, as the width of the hydrogel microgrooves increased, the number of cells with orientation angles <30° gradually decreased (79.9%, 62.3%, 54.7%, respectively), while the number of cells with orientation angles between 60°-90° increased (P<0.001). The cell bodies in the microgrooves were smaller and more rounded in shape. The cells were aligned along the direction of the microgrooves and corresponding changes occurred in the arrangement of the cell cytoskeleton. In the control group, cytoskeletal filaments were aligned in random directions, presenting an orientation angle of (45.5±3.7)°. Cytoskeletal filaments were distributed evenly within various orientation angles. However, in the 60 μm, 100 μm, and 140 μm hydrogel microgrooves, the orientation angles of the cytoskeletal filaments were significantly decreased, measuring (14.4±3.1)°, (24.7±3.5)°, and (31.9±3.3)°, respectively. The number of cytoskeletal filaments with orientation angles <30° significantly increased in hydrogel microgrooves of different widths (P<0.001). However, as the width of the hydrogel microgrooves increased, the number of cytoskeletal filaments with orientation angles <30° gradually decreased, while the number of cytoskeletal filaments with orientation angles between 60°-90° gradually increased (P<0.001). Conclusion Hydrogel microgrooves can regulate the morphology and orientation of HUVECs and mimic to a certain extent the in vivo microenvironment of vascular endothelial cells, providing an experimental model that bears better resemblance to human physiology for the study of the unique physiological functions of vascular endothelial cells. Nonetheless, the molecular mechanism of spatial constraints on the morphology and the assembly of vascular endothelial cell needs to be further investigated.
Collapse
Affiliation(s)
- 文丽 江
- 四川大学华西基础医学与法医学院 生物医学工程研究室 (成都 610041)Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 健 钟
- 四川大学华西基础医学与法医学院 生物医学工程研究室 (成都 610041)Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 智 欧阳
- 四川大学华西基础医学与法医学院 生物医学工程研究室 (成都 610041)Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 钧怡 沈
- 四川大学华西基础医学与法医学院 生物医学工程研究室 (成都 610041)Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 艳 邱
- 四川大学华西基础医学与法医学院 生物医学工程研究室 (成都 610041)Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 烨 曾
- 四川大学华西基础医学与法医学院 生物医学工程研究室 (成都 610041)Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| |
Collapse
|
25
|
Liu K, He X, Zhang Z, Sun T, Chen J, Chen C, Wen W, Ding S, Liu M, Zhou C, Luo B. Highly anisotropic and elastic cellulosic scaffold guiding cell orientation and osteogenic differentiation via topological and mechanical cues. Carbohydr Polym 2023; 321:121292. [PMID: 37739527 DOI: 10.1016/j.carbpol.2023.121292] [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: 05/11/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 09/24/2023]
Abstract
Inspired by the similarity of anisotropic channels in wood to the canals of bone, the elastic wood-derived (EW) scaffolds with anisotropic channels were prepared via simple delignification treatment of natural wood (NW). We hypothesize that the degree of delignification will lead to differences in mechanical properties of scaffolds, which in turn directly affect the behaviors and fate of stem cells. The delignification process did not destroy the anisotropic channel structure of the scaffolds, but endowed the scaffolds with good elasticity and rapid stress relaxation. Interestingly, the micron-scale anisotropic channels of the scaffolds can highly promote the polarization of cells along the direction of channels. We also found that the alkaline phosphatase of EW scaffold can reach to about 13.1 U/gprot, which was about double that of NW scaffold. Moreover, the longer the delignification time, the better the osteogenic activity of the EW scaffolds. We further hypothesize that the osteogenic activity of scaffolds is related to the stress relaxation properties. The immunofluorescence staining showed that when the stress relaxation time of scaffold was shortened to about 10 s, the nuclear ratio of YAP of scaffold increased to 0.22, which well supports our hypothesis.
Collapse
Affiliation(s)
- Kun Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Xiangheng He
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Zhaoyu Zhang
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Tianyi Sun
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Jiaqing Chen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Chunhua Chen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China
| | - Wei Wen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Shan Ding
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Mingxian Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Changren Zhou
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China
| | - Binghong Luo
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, PR China; Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, PR China.
| |
Collapse
|
26
|
Wei Z, Liu J, Jia Y, Lei M, Zhang S, Xi P, Ma Y, Zhang M, Ma J, Wang L, Guo H, Xu F. Fiber Microarchitecture in Interpenetrating Collagen-Alginate Hydrogel with Tunable Mechanical Plasticity Regulates Tumor Cell Migration. Adv Healthc Mater 2023; 12:e2301586. [PMID: 37506713 DOI: 10.1002/adhm.202301586] [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: 05/16/2023] [Revised: 07/13/2023] [Indexed: 07/30/2023]
Abstract
The fiber structures of tumor microenvironment (TME) are well-known in regulating tumor cell behaviors, and the plastic remolding of TME has recently been suggested to enhance tumor metastasis as well. However, the interrelationship between the fiber microarchitecture and matrix plasticity is inextricable by existing in vitro models. The individual roles of fiber microarchitecture and matrix plasticity in tuning tumor cell behaviors remain elusive. This study develops an interpenetrating collagen-alginate hydrogel platform with independently tunable matrix plasticity and fiber microarchitecture through an interpenetrating strategy of alginate networks and collagen I networks. With this hydrogel platform, it is demonstrated that tumor cells in high plasticity hydrogels are more extensive and aggressive than in low plasticity hydrogels and fiber structures only have influence in high plasticity hydrogels. The study further elucidates the underlying mechanisms through analyzing the distribution of forces within the matrix and tracking the focal adhesions (FAs) and finds that highly plastic hydrogels can activate the FAs formation, whereas the maturation and stability of FAs are dominated by fiber dispersion. This study not only establishes new ideas on how cells interact with TME cues but also would help to further finely tailor engineered hydrogel platforms for studying tumor behaviors in vitro.
Collapse
Affiliation(s)
- Zhao Wei
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Jingyi Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yuanbo Jia
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Meng Lei
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Songbai Zhang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, No. 145 West Changle Road, Xi'an, 710032, P. R. China
| | - Pan Xi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yufei Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Min Zhang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, No. 145 West Changle Road, Xi'an, 710032, P. R. China
| | - Jinlu Ma
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Lin Wang
- College of Medicine, Xi'an International University, Xi'an, Shaanxi, 710077, China
- Engineering Research Center of Personalized Anti-aging Health Product Development and Transformation, Universities of Shaanxi Province, Xi'an, Shaanxi, 710077, China
| | - Hui Guo
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
27
|
Liu P, Qiu T, Liu J, Long X, Wang X, Nie H, Yu M, Ma C, Lin N, Teoh SH, Wang Z. Mechanically enhanced and osteobioactive synthetic periosteum via development of poly(ε-caprolactone)/microtantalum composite. Colloids Surf B Biointerfaces 2023; 231:113537. [PMID: 37776773 DOI: 10.1016/j.colsurfb.2023.113537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/02/2023] [Accepted: 09/07/2023] [Indexed: 10/02/2023]
Abstract
Periosteum, the thin layer covering adjacent to bone containing specific architecture, is important for functional bone regeneration and remodeling. Synthetic periosteum investigated presently lacks the resemblance of natural periosteum, suffering from poor mechanical strength and cell attachment. Here, we report a newly-developed biomimetic film to function as synthetic periosteum. Based on poly(ε-caprolactone) (PCL), where surface wettability of the synthetic periosteum is enhanced by microtantalum (mTa) particle blending and after a cold drawing process, further obtains topographical anisotropy without any involvement of solvent. This new blend shows mechanical enhancement over pure PCL, with yield stress and elastic strain approaching the natural periosteum. A distinct degradation mechanism is proposed for the blend, and by seeding with mouse calvarial preosteoblasts, cell proliferation is promoted on surface of the drawn PCL but delayed on the mTa-blended PCL. However, cell mineralization is accelerated on the mTa-blended surface. This is less on the drawn PCL. The synergistical integration of cellular proliferation, alignment and osteogenic enhancement suggest that the cold drawn PCL/Ta blend has unique potential for developing into a synthetic periosteum and other tissue-engineering products.
Collapse
Affiliation(s)
- Peng Liu
- College of Materials Science and Engineering, Hunan University, Changsha 410072, PR China
| | - Tiecheng Qiu
- College of Materials Science and Engineering, Hunan University, Changsha 410072, PR China
| | - Jiabing Liu
- College of Materials Science and Engineering, Hunan University, Changsha 410072, PR China
| | - Xiaoxi Long
- College of Materials Science and Engineering, Hunan University, Changsha 410072, PR China
| | - Xianwei Wang
- Department of Vascular Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Hemin Nie
- College of Biology, Hunan University, Changsha 410072, PR China
| | - Mengqiang Yu
- Department of Neurosurgery, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China.
| | - Chao Ma
- College of Materials Science and Engineering, Hunan University, Changsha 410072, PR China
| | - Nan Lin
- College of Materials Science and Engineering, Hunan University, Changsha 410072, PR China
| | - Swee Hin Teoh
- College of Materials Science and Engineering, Hunan University, Changsha 410072, PR China
| | - Zuyong Wang
- College of Materials Science and Engineering, Hunan University, Changsha 410072, PR China.
| |
Collapse
|
28
|
Guo H, Peng X, Dong X, Li J, Cheng C, Wei Q. Promoting Stem Cell Mechanosensing and Osteogenesis by Hybrid Soft Fibers. ACS APPLIED MATERIALS & INTERFACES 2023; 15:47880-47892. [PMID: 37788009 DOI: 10.1021/acsami.3c07999] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Bone regenerative biomaterials are essential in treating bone defects as they serve as extracellular matrix (ECM) mimics, creating a favorable environment for cell attachment, proliferation, and differentiation. However, the currently used bone regenerative biomaterials mostly exhibit high stiffness, which may lead to difficulties in degradation and thus increase the risk of foreign body ingestion. In this study, we prepared soft fibrous scaffolds modified with Zn-MOF-74 nanoparticles via electrostatic spinning. The soft fibers (only 1 kPa) permit remodeling under cellular adhesive force, optimizing the mechanical cues in the microenvironment to support cell adhesion and mechanosensing. In addition, the incorporation of Zn-MOF-74 nanoparticles enables the stable and sustained release of zinc ions, promoting stem cell mechanotransduction and osteogenic differentiation. Therefore, the hybrid soft fibers facilitate the regeneration of new bone in the rat femoral defect model, which provides a promising approach for regenerative medicine.
Collapse
Affiliation(s)
- Hui Guo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Xu Peng
- West China School of Basic Medical Sciences & Forensic Medicine, Experimental and Research Animal Institute, Sichuan University, Chengdu 610065, China
| | - Xiangyu Dong
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Jiangge Li
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
29
|
Yin B, Zhang Q, Yan J, Huang Y, Li C, Chen J, Wen C, Wong SHD, Yang M. Nanomanipulation of Ligand Nanogeometry Modulates Integrin/Clathrin-Mediated Adhesion and Endocytosis of Stem Cells. NANO LETTERS 2023; 23:9160-9169. [PMID: 37494286 DOI: 10.1021/acs.nanolett.3c01757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Nanosubstrate engineering can be a biomechanical approach for modulating stem cell differentiation in tissue engineering. However, the study of the effect of clathrin-mediated processes on manipulating this behavior is unexplored. Herein, we develop integrin-binding nanosubstrates with confined nanogeometries that regulate clathrin-mediated adhesion- or endocytosis-active signaling pathways for modulating stem fates. Isotropically presenting ligands on the nanoscale enhances the expression of clathrin in cells, thereby facilitating uptake of dexamethasone-loaded nanoparticles (NPs) to boost osteogenesis of stem cells. In contrast, anisotropic ligand nanogeometry suppresses this clathrin-mediated NP entry by strengthening the association between clathrin and adhesion spots to reinforce mechanotransduced signaling, which can be abrogated by the pharmacological inhibition of clathrin. Meanwhile, inhibiting focal adhesion formation hinders cell spreading and enables a higher endocytosis efficiency. Our findings reveal the crucial roles of clathrin in both endocytosis and mechanotransduction of stem cells and provide the parameter of ligand nanogeometry for the rational design of biomaterials for tissue engineering.
Collapse
Affiliation(s)
- Bohan Yin
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Qin Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Jiaxiang Yan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Yingying Huang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Chuanqi Li
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Jiareng Chen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Chunyi Wen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Siu Hong Dexter Wong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| |
Collapse
|
30
|
Bao M, Xie J. Geometric Confinement-Mediated Mechanical Tension Directs Patterned Differentiation of Mouse ESCs into Organized Germ Layers. ACS APPLIED MATERIALS & INTERFACES 2023; 15:34397-34406. [PMID: 37458389 DOI: 10.1021/acsami.3c03798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
The self-organization of embryonic stem cells (ESCs) into organized tissues with three distinct germ layers is critical to morphogenesis and early development. While the regulation of this self-organization by soluble signals is well established, the involvement of mechanical force gradients in this process remains unclear due to the lack of a suitable platform to study this process. In this study, we developed a 3D microenvironment to examine the influence of mechanical tension gradients on ESC-patterned differentiation during morphogenesis by controlling the geometrical signals (shape and size) of ESC colonies. We found that changes in colony geometry impacted the germ layer pattern, with Cdx2-positive cells being more abundant at edges and in areas with high curvatures. The differentiation patterns were determined by geometry-mediated cell tension gradients, with an extraembryonic mesoderm-like layer forming in high-tension regions and ectodermal-like lineages at low-tension regions in the center. Suppression of cytoskeletal tension hindered ESC self-organization. These results indicate that geometric confinement-mediated mechanical tension plays a crucial role in linking multicellular organization to cell differentiation and impacting tissue patterning.
Collapse
Affiliation(s)
- Min Bao
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou 325001, Zhejiang, China
| | - Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
31
|
Su BY, Chen ZJ, Lv JC, Wang ZG, Huang FW, Liu Y, Luo E, Wang J, Xu JZ, Li ZM. Scalable Fabrication of Polymeric Composite Microspheres to Inhibit Oral Pathogens and Promote Osteogenic Differentiation of Periodontal Membrane Stem Cells. ACS Biomater Sci Eng 2023; 9:4431-4441. [PMID: 37452570 DOI: 10.1021/acsbiomaterials.3c00452] [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: 07/18/2023]
Abstract
Periodontitis is a worldwide bacterial infectious disease, resulting in the resorption of tooth-supporting structures. Biodegradable polymeric microspheres are emerging as an appealing local therapy candidate for periodontal defect regeneration but suffer from tedious procedures and low yields. Herein, we developed a facile yet scalable approach to prepare polylactide composite microspheres with outstanding drug-loading capability. It was realized by blending equimolar polylactide enantiomers at the temperature between the melting point of homocrystallites and stereocomplex (sc) crystallites, enabling the precipitation of sc crystallites in the form of microspheres. Meanwhile, epigallocatechin gallate (EGCG) and nano-hydroxyapatite were encapsulated in the microspheres in the designated amount. Such an assembly allowed the fast and sustained release of EGCG and Ca2+ ions. The resultant hybrid composite microspheres not only exhibited strong antimicrobial activity against typical oral pathogens (Porphyromonas gingivalis and Enterococcus faecalis), but also directly promoted osteogenic differentiation of periodontal ligament stem cells with good cytocompatibility. These dual-functional composite microspheres offer a desired drug delivery platform to address the practical needs for periodontitis treatment.
Collapse
Affiliation(s)
- Biao-Yao Su
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Zi-Jian Chen
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jia-Cheng Lv
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Zhi-Guo Wang
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fu-Wen Huang
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Jing Wang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jia-Zhuang Xu
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Zhong-Ming Li
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
32
|
Joshi A, Kaur T, Singh N. Exploiting the Biophysical Cues toward Dual Differentiation of hMSC's within Geometrical Patterns. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:6691-6697. [PMID: 37129583 DOI: 10.1021/acs.langmuir.3c00070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
A wide variety of cues from the extracellular matrix (ECM) have been known to affect the differentiation of stem cells in vivo. In particular, the biophysical cues and cell shape have been known to affect the stem cell function, yet very little is known about the interplay between how these cues control differentiation. For the first time, by using photolithography to pattern poly(ethylene glycol) (PEG), patterns of square and triangular geometries were created, and the effect of these structures and the biophysical cues arising were utilized to differentiate cells into multiple lineages inside a same pattern without the use of any adhered protein or growth factors. The data from these studies showed that the cells present at the edges were well elongated, exhibit high aspect ratios, and differentiated into osteogenic lineage, whereas the cells present at the center exhibit lower aspect ratio and were primarily adipogenic lineage regardless of the geometry. This was correlated to the higher expression of focal adhesion proteins at the edges, the expression of which have been known to affect the osteogenic differentiation. By showing MSC lineage commitment relationships due to physical signals, this study highlights the importance of these cues and cell shape in further understanding stem cell behavior for tissue engineering applications.
Collapse
Affiliation(s)
- Akshay Joshi
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Tejinder Kaur
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
33
|
Zhou Z, Zhang J, He X, Chen X, Dong L, Lin J, Wang H, Weng W, Cheng K. Regulation of Macrophage Polarization on Chiral Potential Distribution of CFO/P(VDF-TrFE) Films. ACS Biomater Sci Eng 2023; 9:2524-2533. [PMID: 37092816 DOI: 10.1021/acsbiomaterials.3c00207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Surface potentials of biomaterials have been shown to regulate cell fate commitment. However, the effects of chirality-patterned potential distribution on macrophage polarization are still only beginning to be explored. In this work, we demonstrated that the chirality-patterned potential distribution of CoFe2O4/poly(vinylidene fluoride-trifluoroethylene) (CFO/P(VDF-TrFE)) films could significantly down-regulate the M1 polarization of bone marrow-derived macrophages (BMDMs). Specifically, the dextral-patterned surface potential distribution simultaneously up-regulated the expression of M2-related markers of BMDMs. The results were attributed to the sensitive difference of integrin subunits (α5β1 and αvβ3) to the dextral- and sinistral-patterned surface potential distribution, respectively. The interaction difference between the integrin subunits and surface potential distribution altered the cell adhesion and cytoskeletal structure and thereby the polarization behavior of BMDMs. This work, therefore, emphasizes the importance of chirality of potential distribution on cell behavior and provides a new strategy to regulate the immune response of biomaterials.
Collapse
Affiliation(s)
- Zhiyuan Zhou
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| | - Jiamin Zhang
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| | - Xuzhao He
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| | - Xiaoyi Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Lingqing Dong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Jun Lin
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Province Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Wenjian Weng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| | - Kui Cheng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
34
|
Lei X, Miao S, Wang X, Gao Y, Wu H, Cheng P, Song Y, Bi L, Pei G. Microgroove Cues Guiding Fibrogenesis of Stem Cells via Intracellular Force. ACS APPLIED MATERIALS & INTERFACES 2023; 15:16380-16393. [PMID: 36961871 DOI: 10.1021/acsami.2c20903] [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: 06/18/2023]
Abstract
Groove patterns are widely used in material surface modifications. However, the independent role of ditches/ridges in regulating fibrosis of soft tissues is not well-understood, especially the lack of linkage evidence in vitro and in vivo. Herein, two kinds of combinational microgroove chips with the gradient ditch/ridge width were fabricated by photolithography technology, termed R and G groups, respectively. In group R, the ridge width was 1, 5, 10, and 30 μm, with a ditch width of 30 μm; in group G, the groove width was 5, 10, 20, and 30 μm, and the ridge width was 5 μm. The effect of microgrooves on the morphology, proliferation, and expression of fibrous markers of stem cells was systematically investigated in vitro. Moreover, thicknesses of fibrous capsules were evaluated after chips were implanted into the muscular pouches of rats for 5 months. The results show that microgrooves have almost no effect on cell proliferation but significantly modulate the morphology of cells and focal adhesions (FAs) in vitro, as well as fibrosis differentiation. In particular, the differentiation of stem cells is attenuated after the intracellular force caused by stress fibers and FAs is interfered by drugs, such as rotenone and blebbistatin. Histological analysis shows that patterns of high intracellular force can apparently stimulate soft tissue fibrosis in vivo. This study not only reveals the specific rules and mechanisms of ditch/ridge regulating stem cell behaviors but also offers insight into tailoring implant surface patterns to induce controlled soft tissue fibrosis.
Collapse
Affiliation(s)
- Xing Lei
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
- Department of Orthopedic Surgery, Linyi People's Hospital, Linyi 276000, China
| | - Sheng Miao
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Xiuli Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Yi Gao
- Southern University of Science and Technology Hospital, No. 6019 Liuxian Street, Xili Avenue, Nanshan District, Shenzhen 518055, China
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hao Wu
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Pengzhen Cheng
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Yue Song
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Long Bi
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
| | - Guoxian Pei
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an 710032, China
- Southern University of Science and Technology Hospital, No. 6019 Liuxian Street, Xili Avenue, Nanshan District, Shenzhen 518055, China
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
35
|
Liu S, Yu JM, Gan YC, Qiu XZ, Gao ZC, Wang H, Chen SX, Xiong Y, Liu GH, Lin SE, McCarthy A, John JV, Wei DX, Hou HH. Biomimetic natural biomaterials for tissue engineering and regenerative medicine: new biosynthesis methods, recent advances, and emerging applications. Mil Med Res 2023; 10:16. [PMID: 36978167 PMCID: PMC10047482 DOI: 10.1186/s40779-023-00448-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/23/2023] [Indexed: 03/30/2023] Open
Abstract
Biomimetic materials have emerged as attractive and competitive alternatives for tissue engineering (TE) and regenerative medicine. In contrast to conventional biomaterials or synthetic materials, biomimetic scaffolds based on natural biomaterial can offer cells a broad spectrum of biochemical and biophysical cues that mimic the in vivo extracellular matrix (ECM). Additionally, such materials have mechanical adaptability, microstructure interconnectivity, and inherent bioactivity, making them ideal for the design of living implants for specific applications in TE and regenerative medicine. This paper provides an overview for recent progress of biomimetic natural biomaterials (BNBMs), including advances in their preparation, functionality, potential applications and future challenges. We highlight recent advances in the fabrication of BNBMs and outline general strategies for functionalizing and tailoring the BNBMs with various biological and physicochemical characteristics of native ECM. Moreover, we offer an overview of recent key advances in the functionalization and applications of versatile BNBMs for TE applications. Finally, we conclude by offering our perspective on open challenges and future developments in this rapidly-evolving field.
Collapse
Affiliation(s)
- Shuai Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900 China
| | - Jiang-Ming Yu
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336 China
| | - Yan-Chang Gan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900 China
| | - Xiao-Zhong Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900 China
| | - Zhe-Chen Gao
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336 China
| | - Huan Wang
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033 Guangdong China
| | - Shi-Xuan Chen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325011 Zhejiang China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Guo-Hui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Si-En Lin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong SAR, 999077 China
| | - Alec McCarthy
- Department of Functional Materials, Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Johnson V. John
- Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68130 USA
| | - Dai-Xu Wei
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336 China
- Zigong Affiliated Hospital of Southwest Medical University, Zigong Psychiatric Research Center, Zigong Institute of Brain Science, Zigong, 643002 Sichuan China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710127 China
| | - Hong-Hao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900 China
| |
Collapse
|
36
|
Topographical cues of PLGA membranes modulate the behavior of hMSCs, myoblasts and neuronal cells. Colloids Surf B Biointerfaces 2023; 222:113070. [PMID: 36495697 DOI: 10.1016/j.colsurfb.2022.113070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/11/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Biomaterial surface modification through the introduction of defined and repeated patterns of topography helps study cell behavior in response to defined geometrical cues. The lithographic molding technique is widely used for conferring biomaterial surface microscale cues and enhancing the performance of biomedical devices. In this work, different master molds made by UV mask lithography were used to prepare poly (D,L-lactide-co-glycolide) - PLGA micropatterned membranes to present different features of topography at the cellular interface: channels, circular pillars, rectangular pillars, and pits. The effects of geometrical cues were investigated on different cell sources, such as neuronal cells, myoblasts, and stem cells. Morphological evaluation revealed a peculiar cell arrangement in response to a specific topographical stimulus sensed over the membrane surface. Cells seeded on linear-grooved membranes showed that this cue promoted elongated cell morphology. Rectangular and circular pillars act instead as discontinuous cues at the cell-membrane interface, inducing cell growth in multiple directions. The array of pits over the surface also highlighted the precise spatiotemporal organization of the cell; they grew between the interconnected membrane space within the pits, avoiding the microscale hole. The overall approach allowed the evaluation of the responses of different cell types adhered to various surface patterns, build-up on the same polymeric membrane, and disclosing the effect of specific topographical features. We explored how various microtopographic signals play distinct roles in different cells, thus affecting cell adhesion, migration, differentiation, cell-cell interactions, and other metabolic activities.
Collapse
|
37
|
Xin H, Shi Q, Ning X, Chen Y, Jia X, Zhang Z, Zhu S, Li Y, Liu F, Kong L. Biomimetic Mineralized Fiber Bundle-Inspired Scaffolding Surface on Polyetheretherketone Implants Promotes Osseointegration. Macromol Biosci 2023; 23:e2200436. [PMID: 36617598 DOI: 10.1002/mabi.202200436] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/11/2022] [Indexed: 01/10/2023]
Abstract
The stress shielding effect caused by traditional metal implants is circumvented by using polyetheretherketone (PEEK), due to its excellent mechanical properties; however, the biologically inert nature of PEEK limits its application. Endowing PEEK with biological activity to promote osseointegration would increase its applicability for bone replacement implants. A biomimetic study is performed, inspired by mineralized collagen fiber bundles that contact bone marrow mesenchymal stem cells (BMMSCs) on the native trabecular bone surface. The PEEK surface (P) is first sulfonated with sulfuric acid to form a porous network structure (sP). The surface is then encapsulated with amorphous hydroxyapatite (HA) by magnetron sputtering to form a biomimetic scaffold that resembles mineralized collagen fiber bundles (sPHA). Amorphous HA simulates the composition of osteogenic regions in vivo and exhibits strong biological activity. In vitro results show that more favorable cell adhesion and osteogenic differentiation can be attained with the novelsurface of sPHA than with SP. The results of in vivo experiments show that sPHA exhibits osteoinductive and osteoconductive activity and facilitates bone formation and osseointegration. Therefore, the surface modification strategy can significantly improve the biological activity of PEEK, facilitate effective osseointegration, and inspire further bionic modification of other inert polymers similar to PEEK.
Collapse
Affiliation(s)
- He Xin
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Qianwen Shi
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaona Ning
- Department of Ophthalmology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Yicheng Chen
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xuelian Jia
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.,College of Life Sciences, Northwest University, Xi'an, 710032, China
| | - Zhouyang Zhang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Simin Zhu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.,College of Life Sciences, Northwest University, Xi'an, 710032, China
| | - Yunpeng Li
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Fuwei Liu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Liang Kong
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
38
|
Sun Q, Pei F, Zhang M, Zhang B, Jin Y, Zhao Z, Wei Q. Curved Nanofiber Network Induces Cellular Bridge Formation to Promote Stem Cell Mechanotransduction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204479. [PMID: 36382560 PMCID: PMC9875655 DOI: 10.1002/advs.202204479] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Remarkable exertions are directed to reveal and understand topographic cues that induce cell mechanical sensitive responses including lineage determination. Extracellular matrix (ECM) is the sophisticated ensemble of diverse factors offering the complicated cellular microenvironment to regulate cell behaviors. However, the functions of only a few of these factors are revealed; most of them are still poorly understood. Herein, the focus is on understanding the curved structure in ECM network for regulating stem cell mechanotransduction. A curved nanofiber network mimicking the curved structure in ECM is fabricated by an improved electrospinning technology. Compared with the straight fibers, the curved fibers promote cell bridge formation because of the cytoskeleton tension. The actomyosin filaments are condensed near the curved edge of the non-adhesive bridge in the bridging cells, which generates higher myosin-II-based intracellular force. This force drives cell lineage commitment toward osteogenic differentiation. This study enriches and perfects the knowledge of the effects of topographic cues on cell behaviors and guides the development of novel biomaterials.
Collapse
Affiliation(s)
- Qian Sun
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065P. R. China
| | - Fang Pei
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065P. R. China
| | - Man Zhang
- College of Biomedical EngineeringSichuan UniversityChengdu610065P. R. China
| | - Bo Zhang
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Ying Jin
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Zhihe Zhao
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Qiang Wei
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065P. R. China
- College of Biomedical EngineeringSichuan UniversityChengdu610065P. R. China
| |
Collapse
|
39
|
Zhang H, Ma Y, Wang Y, Niu L, Zou R, Zhang M, Liu H, Genin GM, Li A, Xu F. Rational Design of Soft-Hard Interfaces through Bioinspired Engineering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204498. [PMID: 36228093 DOI: 10.1002/smll.202204498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Soft-hard tissue interfaces in nature present a diversity of hierarchical transitions in composition and structure to address the challenge of stress concentrations that would otherwise arise at their interface. The translation of these into engineered materials holds promise for improved function of biomedical interfaces. Here, soft-hard tissue interfaces found in the body in health and disease, and the application of the diverse, functionally graded, and hierarchical structures that they present to bioinspired engineering materials are reviewed. A range of such bioinspired engineering materials and associated manufacturing technologies that are on the horizon in interfacial tissue engineering, hydrogel bioadhesion at the interfaces, and healthcare and medical devices are described.
Collapse
Affiliation(s)
- Hui Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yufei Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yijie Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Lin Niu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Rui Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Min Zhang
- State Key Laboratory of Military Stomatology, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Hao Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Guy M Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, 63130, USA
- NSF Science and Technology Center for Engineering MechanoBiology, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
40
|
Huang P, Yang P, Liu K, Tao W, Tao J, Ai F. Evaluation of 'surgery-friendly' bone scaffold characteristics: 3D printed ductile BG/PCL scaffold with high inorganic content to repair critical bone defects. Biomed Mater 2022; 18. [PMID: 36317271 DOI: 10.1088/1748-605x/ac9e34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/27/2022] [Indexed: 12/23/2022]
Abstract
The repair of irregular and complex critical bone defects remains a challenge in clinical practice. The application of 3D-printed bioceramics particle/polymer composite scaffolds in bone tissue engineering has been widely studied. At present, the inorganic particle content of the composite scaffolds is generally low, resulting in poor osteogenic activity. However, scaffold with high inorganic content are highly brittle, difficult to operate during surgery, and cannot be in close contact with surrounding bones. Therefore, it is of great significance to design a 'surgery-friendly' scaffold with high bioceramic content and good ductility. In this study, we used the solvent method to add high concentration (wt% 70%) bioglass (BG) into polycaprolactone (PCL), and polyethylene glycol was used as plasticizer to prepare 70% BG/PCL composite scaffolds with high ductility using 3D printing technology.In vitroexperiments showed that the scaffold had good mechanical properties: easy extension, easy folding and strong compressive resistance. It also showed good performance in biocompatibility and osteogenic activity. It was further observed that compared with pure BG or PCL implantation, the scaffold with higher BG content could have more new bone tissue appeared after 12 weeks. All these results indicate that 3D-printed 70% BG/PCL scaffolds have great potential for personalized repair of bone defects.
Collapse
Affiliation(s)
- Pengren Huang
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Peng Yang
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Keming Liu
- Department of Orthopaedics, Guixi Dongxin Hospital, Yingtan 335400, People's Republic of China
| | - Wei Tao
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Jun Tao
- Department of Orthopaedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Fanrong Ai
- School of Advanced Manufacturing, Nanchang University, Nanchang 330031, People's Republic of China
| |
Collapse
|
41
|
Cao D, Ding J. Recent advances in regenerative biomaterials. Regen Biomater 2022; 9:rbac098. [PMID: 36518879 PMCID: PMC9745784 DOI: 10.1093/rb/rbac098] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 07/22/2023] Open
Abstract
Nowadays, biomaterials have evolved from the inert supports or functional substitutes to the bioactive materials able to trigger or promote the regenerative potential of tissues. The interdisciplinary progress has broadened the definition of 'biomaterials', and a typical new insight is the concept of tissue induction biomaterials. The term 'regenerative biomaterials' and thus the contents of this article are relevant to yet beyond tissue induction biomaterials. This review summarizes the recent progress of medical materials including metals, ceramics, hydrogels, other polymers and bio-derived materials. As the application aspects are concerned, this article introduces regenerative biomaterials for bone and cartilage regeneration, cardiovascular repair, 3D bioprinting, wound healing and medical cosmetology. Cell-biomaterial interactions are highlighted. Since the global pandemic of coronavirus disease 2019, the review particularly mentions biomaterials for public health emergency. In the last section, perspectives are suggested: (i) creation of new materials is the source of innovation; (ii) modification of existing materials is an effective strategy for performance improvement; (iii) biomaterial degradation and tissue regeneration are required to be harmonious with each other; (iv) host responses can significantly influence the clinical outcomes; (v) the long-term outcomes should be paid more attention to; (vi) the noninvasive approaches for monitoring in vivo dynamic evolution are required to be developed; (vii) public health emergencies call for more research and development of biomaterials; and (viii) clinical translation needs to be pushed forward in a full-chain way. In the future, more new insights are expected to be shed into the brilliant field-regenerative biomaterials.
Collapse
Affiliation(s)
- Dinglingge Cao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| |
Collapse
|
42
|
Zhu M, Zhang R, Mao Z, Fang J, Ren F. Topographical biointerface regulating cellular functions for bone tissue engineering. BIOSURFACE AND BIOTRIBOLOGY 2022; 8:165-187. [DOI: 10.1049/bsb2.12043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/06/2022] [Indexed: 03/01/2025] Open
Abstract
AbstractThe physiochemical properties of the implant interface significantly influence cell growth, differentiation, cellular matrix deposition, and mineralisation, and eventually, determine the bone regeneration efficiency. Cells directly sense and respond to the physical, chemical, and mechanical cues of the implant surface, and it is increasingly recognized that surface topography can evoke specific cellular responses, conferring biological functions on substrate materials and regulating tissue regeneration. Current progress towards the fundamental understanding of the interplay between the cell and topographical surface has been made by combined advance in fabrication technologies and cell biology. Particularly, the precise fabrication and control of nano/microscale topographies can provide the fundamental knowledge of the mechanotransduction process that governs the cellular response as well as the knowledge of how the specific features drive cells towards a defined differentiation outcome. In this review, we first introduce common techniques and substrate materials for designing and fabricating micro/nano‐topographical surfaces for bone regeneration. We then illustrate the intrinsic relationship of topological cues, cellular signal transduction, and cell functions and fates in osteogenic differentiation. Finally, we discuss the challenges and the future of using topological cues as a cell therapy to direct bone tissue regeneration.
Collapse
Affiliation(s)
- Mingyu Zhu
- Department of Materials Science and Engineering Southern University of Science and Technology Shenzhen Guangdong China
| | - Rui Zhang
- Department of Prosthodontics Stomatology Center Peking University Shenzhen Hospital Shenzhen Guangdong China
| | - Zhixiang Mao
- Department of Materials Science and Engineering Southern University of Science and Technology Shenzhen Guangdong China
| | - Ju Fang
- Department of Materials Science and Engineering Southern University of Science and Technology Shenzhen Guangdong China
| | - Fuzeng Ren
- Department of Materials Science and Engineering Southern University of Science and Technology Shenzhen Guangdong China
| |
Collapse
|
43
|
Hsu CC, Serio A, Gopal S, Gelmi A, Chiappini C, Desai RA, Stevens MM. Biophysical Regulations of Epigenetic State and Notch Signaling in Neural Development Using Microgroove Substrates. ACS APPLIED MATERIALS & INTERFACES 2022; 14:32773-32787. [PMID: 35830496 PMCID: PMC9335410 DOI: 10.1021/acsami.2c01996] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A number of studies have recently shown how surface topography can alter the behavior and differentiation patterns of different types of stem cells. Although the exact mechanisms and molecular pathways involved remain unclear, a consistent portion of the literature points to epigenetic changes induced by nuclear remodeling. In this study, we investigate the behavior of clinically relevant neural populations derived from human pluripotent stem cells when cultured on polydimethylsiloxane microgrooves (3 and 10 μm depth grooves) to investigate what mechanisms are responsible for their differentiation capacity and functional behavior. Our results show that microgrooves enhance cell alignment, modify nuclear geometry, and significantly increase cellular stiffness, which we were able to measure at high resolution with a combination of light and electron microscopy, scanning ion conductance microscopy (SICM), and atomic force microscopy (AFM) coupled with quantitative image analysis. The microgrooves promoted significant changes in the epigenetic landscape, as revealed by the expression of key histone modification markers. The main behavioral change of neural stem cells on microgrooves was an increase of neuronal differentiation under basal conditions on the microgrooves. Through measurements of cleaved Notch1 levels, we found that microgrooves downregulate Notch signaling. We in fact propose that microgroove topography affects the differentiation potential of neural stem cells by indirectly altering Notch signaling through geometric segregation and that this mechanism in parallel with topography-dependent epigenetic modulations acts in concert to enhance stem cell neuronal differentiation.
Collapse
Affiliation(s)
- Chia-Chen Hsu
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Andrea Serio
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Sahana Gopal
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Amy Gelmi
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Ciro Chiappini
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Ravi A. Desai
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Molly M. Stevens
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| |
Collapse
|
44
|
Bubble-Patterned Films by Inkjet Printing and Gas Foaming. COATINGS 2022. [DOI: 10.3390/coatings12060806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The micropatterning of thin films represents a challenging task, even for additive manufacturing techniques. In this work, we introduce the use of inkjet-printing technology coupled with a gas-foaming process, to produce patterned porosities on polymeric thin films, to develop a bubble-writing method. Inkjet printing of an aqueous solution of poly (vinyl alcohol) (PVA), a well-known gas-barrier polymer, allows the selective coating of a thin poly (lactic acid) (PLA) film, which is, successively, exposed to a gas-foaming process. The foaming of the thin PLA film is effective, only when PVA is printed on top, since the PVA barrier hinders the premature loss of the gas, thus allowing the formation of cavities (bubbles) in the covered areas; then, removing the PVA coating by water washing forms a bubble pattern. As a proof of concept, the surface-morphology features of the patterned porous PLA films have been proven effective at driving endothelial cell growth. A new technological platform is, hence, introduced in the field of tissue engineering and, in general, in fields involving thin films, where a patterned porous structure may add value.
Collapse
|
45
|
Lin C, Zheng X, Lin S, Zhang Y, Wu J, Li Y. Mechanotransduction Regulates the Interplays Between Alveolar Epithelial and Vascular Endothelial Cells in Lung. Front Physiol 2022; 13:818394. [PMID: 35250619 PMCID: PMC8895143 DOI: 10.3389/fphys.2022.818394] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/28/2022] [Indexed: 12/22/2022] Open
Abstract
Mechanical stress plays a critical role among development, functional maturation, and pathogenesis of pulmonary tissues, especially for the alveolar epithelial cells and vascular endothelial cells located in the microenvironment established with vascular network and bronchial-alveolar network. Alveolar epithelial cells are mainly loaded by cyclic strain and air pressure tension. While vascular endothelial cells are exposed to shear stress and cyclic strain. Currently, the emerging evidences demonstrated that non-physiological mechanical forces would lead to several pulmonary diseases, including pulmonary hypertension, fibrosis, and ventilation induced lung injury. Furthermore, a series of intracellular signaling had been identified to be involved in mechanotransduction and participated in regulating the physiological homeostasis and pathophysiological process. Besides, the communications between alveolar epithelium and vascular endothelium under non-physiological stress contribute to the remodeling of the pulmonary micro-environment in collaboration, including hypoxia induced injuries, endothelial permeability impairment, extracellular matrix stiffness elevation, metabolic alternation, and inflammation activation. In this review, we aim to summarize the current understandings of mechanotransduction on the relation between mechanical forces acting on the lung and biological response in mechanical overloading related diseases. We also would like to emphasize the interplays between alveolar epithelium and vascular endothelium, providing new insights into pulmonary diseases pathogenesis, and potential targets for therapy.
Collapse
Affiliation(s)
- Chuyang Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaolan Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Sha Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yue Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jinlin Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
46
|
Zhao T, Zhang J, Gao X, Yuan D, Gu Z, Xu Y. Electrospun Nanofibers for Bone Regeneration: From Biomimetic Composition, Structure to Function. J Mater Chem B 2022; 10:6078-6106. [DOI: 10.1039/d2tb01182d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In recent years, a variety of novel materials and processing technologies have been developed to prepare tissue engineering scaffolds for bone defect repair. Among them, nanofibers fabricated via electrospinning technology...
Collapse
|