1
|
Zhang W, Rao Y, Wong SH, Wu Y, Zhang Y, Yang R, Tsui SK, Ker DFE, Mao C, Frith JE, Cao Q, Tuan RS, Wang DM. Transcriptome-Optimized Hydrogel Design of a Stem Cell Niche for Enhanced Tendon Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2313722. [PMID: 39417770 PMCID: PMC11733723 DOI: 10.1002/adma.202313722] [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: 12/15/2023] [Revised: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Bioactive hydrogels have emerged as promising artificial niches for enhancing stem cell-mediated tendon repair. However, a substantial knowledge gap remains regarding the optimal combination of niche features for targeted cellular responses, which often leads to lengthy development cycles and uncontrolled healing outcomes. To address this critical gap, an innovative, data-driven materiomics strategy is developed. This approach is based on in-house RNA-seq data that integrates bioinformatics and mathematical modeling, which is a significant departure from traditional trial-and-error methods. It aims to provide both mechanistic insights and quantitative assessments and predictions of the tenogenic effects of adipose-derived stem cells induced by systematically modulated features of a tendon-mimetic hydrogel (TenoGel). The knowledge generated has enabled a rational approach for TenoGel design, addressing key considerations, such as tendon extracellular matrix concentration, uniaxial tensile loading, and in vitro pre-conditioning duration. Remarkably, our optimized TenoGel demonstrated robust tenogenesis in vitro and facilitated tendon regeneration while preventing undesired ectopic ossification in a rat tendon injury model. These findings shed light on the importance of tailoring hydrogel features for efficient tendon repair. They also highlight the tremendous potential of the innovative materiomics strategy as a powerful predictive and assessment tool in biomaterial development for regenerative medicine.
Collapse
Affiliation(s)
- Wanqi Zhang
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Ying Rao
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Shing Hei Wong
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Hong Kong Bioinformatics CentreThe Chinese University of Hong KongHong Kong SARChina
| | - Yeung Wu
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Yuanhao Zhang
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Rui Yang
- Department of Sports MedicineOrthopedicsSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
| | - Stephen Kwok‐Wing Tsui
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Hong Kong Bioinformatics CentreThe Chinese University of Hong KongHong Kong SARChina
| | - Dai Fei Elmer Ker
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SARChina
- Department of Orthopaedics and TraumatologyFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| | - Chuanbin Mao
- Department of Biomedical EngineeringThe Chinese University of Hong KongHong Kong SARChina
| | - Jessica E. Frith
- Materials Science and EngineeringMonash UniversityClayton3800VICAustralia
- Australian Regenerative Medicine InstituteMonash UniversityClayton3800VICAustralia
- Australian Research Council Training Centre for Cell and Tissue Engineering TechnologiesMonash UniversityClayton3800VICAustralia
| | - Qin Cao
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Hong Kong Bioinformatics CentreThe Chinese University of Hong KongHong Kong SARChina
| | - Rocky S. Tuan
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SARChina
- Department of Orthopaedics and TraumatologyFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| | - Dan Michelle Wang
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SARChina
- Department of Orthopaedics and TraumatologyFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| |
Collapse
|
2
|
Zhou W, Lin J, Wang Q, Wang X, Yao X, Yan Y, Sun W, Zhu Q, Zhang X, Wang X, Ji B, Ouyang H. Chromatin-site-specific accessibility: A microtopography-regulated door into the stem cell fate. Cell Rep 2024; 44:115106. [PMID: 39723890 DOI: 10.1016/j.celrep.2024.115106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 11/12/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Biomaterials that mimic extracellular matrix topography are crucial in tissue engineering. Previous research indicates that certain biomimetic topography can guide stem cells toward multiple specific lineages. However, the mechanisms by which topographic cues direct stem cell differentiation remain unclear. Here, we demonstrate that microtopography influences nuclear tension in mesenchymal stem cells (MSCs), shaping chromatin accessibility and determining lineage commitment. On aligned substrates, MSCs exhibit high cytoskeletal tension along the fiber direction, creating anisotropic nuclear stress that opens chromatin sites for neurogenic, myogenic, and tenogenic genes via transcription factors like Nuclear receptor TLX (TLX). In contrast, random substrates induce isotropic nuclear stress, promoting chromatin accessibility for osteogenic and chondrogenic genes through Runt-related transcription factors (RUNX). Our findings reveal that aligned and random microtopographies direct site-specific chromatin stretch and lineage-specific gene expression, priming MSCs for distinct lineages. This study introduces a novel framework for understanding how topographic cues govern cell fate in tissue repair and regeneration.
Collapse
Affiliation(s)
- Wenyan Zhou
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; School of Medicine, Taizhou University, Taizhou, Zhejiang Province 318000, China
| | - Junxin Lin
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; School of Medicine, Taizhou University, Taizhou, Zhejiang Province 318000, China
| | - Qianchun Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325000, China
| | - Xianliu Wang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 200051, China
| | - Xudong Yao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province 322000, China
| | - Yiyang Yan
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang Province 314400, China
| | - Wei Sun
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang Province 314400, China
| | - Qiuwen Zhu
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Xiaoan Zhang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Xiaozhao Wang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Baohua Ji
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, Zhejiang Province 310027, China; Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310027, China.
| | - Hongwei Ouyang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang Province 314400, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang Province 310058, China.
| |
Collapse
|
3
|
Yan S, Lu Y, An C, Hu W, Chen Y, Li Z, Wei W, Chen Z, Zeng X, Xu W, Lv Z, Pan F, Gao W, Wu Y. Biomechanical research using advanced micro-nano devices: In-Vitro cell Characterization focus. J Adv Res 2024:S2090-1232(24)00602-7. [PMID: 39701378 DOI: 10.1016/j.jare.2024.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Cells in the body reside in a dynamic microenvironment subjected to various physical stimuli, where mechanical stimulation plays a crucial role in regulating cellular physiological behaviors and functions. AIM OF REVIEW Investigating the mechanisms and interactions of mechanical transmission is essential for understanding the physiological and functional interplay between cells and physical stimuli. Therefore, establishing an in vitro biomechanical stimulation cell culture system holds significant importance for research related to cellular biomechanics. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, we primarily focused on various biomechanically relevant cell culture systems and highlighted the advancements and prospects in their preparation processes. Firstly, we discussed the types and characteristics of biomechanics present in the microenvironment within the human body. Subsequently, we introduced the research progress, working principles, preparation processes, potential advantages, applications, and challenges of various biomechanically relevant in vitro cell culture systems. Additionally, we summarized and categorized currently commercialized biomechanically relevant cell culture systems, offering a comprehensive reference for researchers in related fields.
Collapse
Affiliation(s)
- Shiqiang Yan
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China; Center of Cancer Immunology, Shenzhen Institute of Advanced Technology Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Yan Lu
- Department of Otolaryngology Head & Neck Surgery, The First Hospital, Jinzhou Medical University, Jinzhou 121001, Liaoning, China; Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Changming An
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wanglai Hu
- Translational Research Institute, People's Hospital of Zhengzhou University, Academy of Medical Science, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Yaofeng Chen
- Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Ziwen Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Wenbo Wei
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
| | - Zongzheng Chen
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
| | - Xianhai Zeng
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China
| | - Wei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, Shandong, China
| | - Zhenghua Lv
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, Shandong, China.
| | - Fan Pan
- Center of Cancer Immunology, Shenzhen Institute of Advanced Technology Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China.
| | - Wei Gao
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China.
| | - Yongyan Wu
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China; Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, Guangdong, China.
| |
Collapse
|
4
|
Xiang JY, Kang L, Li ZM, Tseng SL, Wang LQ, Li TH, Li ZJ, Huang JZ, Yu NZ, Long X. Biological scaffold as potential platforms for stem cells: Current development and applications in wound healing. World J Stem Cells 2024; 16:334-352. [PMID: 38690516 PMCID: PMC11056631 DOI: 10.4252/wjsc.v16.i4.334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/20/2024] [Accepted: 03/12/2024] [Indexed: 04/25/2024] Open
Abstract
Wound repair is a complex challenge for both clinical practitioners and researchers. Conventional approaches for wound repair have several limitations. Stem cell-based therapy has emerged as a novel strategy to address this issue, exhibiting significant potential for enhancing wound healing rates, improving wound quality, and promoting skin regeneration. However, the use of stem cells in skin regeneration presents several challenges. Recently, stem cells and biomaterials have been identified as crucial components of the wound-healing process. Combination therapy involving the development of biocompatible scaffolds, accompanying cells, multiple biological factors, and structures resembling the natural extracellular matrix (ECM) has gained considerable attention. Biological scaffolds encompass a range of biomaterials that serve as platforms for seeding stem cells, providing them with an environment conducive to growth, similar to that of the ECM. These scaffolds facilitate the delivery and application of stem cells for tissue regeneration and wound healing. This article provides a comprehensive review of the current developments and applications of biological scaffolds for stem cells in wound healing, emphasizing their capacity to facilitate stem cell adhesion, proliferation, differentiation, and paracrine functions. Additionally, we identify the pivotal characteristics of the scaffolds that contribute to enhanced cellular activity.
Collapse
Affiliation(s)
- Jie-Yu Xiang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lin Kang
- Biomedical Engineering Facility, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Zi-Ming Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Song-Lu Tseng
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Li-Quan Wang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Tian-Hao Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhu-Jun Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jiu-Zuo Huang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Nan-Ze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
5
|
Kurpanik R, Gajek M, Gryń K, Jeleń P, Ścisłowska-Czarnecka A, Stodolak-Zych E. Multiscale characterization of electrospun non-wovens for corneal regeneration: Impact of microstructure on mechanical, optical and biological properties. J Mech Behav Biomed Mater 2024; 152:106437. [PMID: 38354568 DOI: 10.1016/j.jmbbm.2024.106437] [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: 12/08/2023] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/16/2024]
Abstract
The multiscale approach in designing substrates for regenerative medicine endows them with beneficial properties determining their performance in the body. Substrates for corneal regeneration should reveal the proper transparency, mechanical properties and microstructure to maintain the functionality of the regenerated tissue. In our study, series of non-wovens with different fibres orientation (random (R), aligned (A)), topography (shish-kebab (KK), core-shell (CS)) and thickness were fabricated via electrospinning. The samples were assessed for mechanical (static tensile test) and optical properties (spectroscopy UV-Vis). The research evaluated the impact of different microstructures on the viability and morphology of three cell lines (Hs 680, HaCaT and RAW 264.7). The results showed how the fibres arrangement influenced mechanical behaviour of the non-wovens. The randomly oriented fibres were more elongated (up to 50 mm) and had a lower maximum tensile force (up to 0.46 N). In turn, the aligned fibres were characterized by lower elongation (up to 19 mm) and higher force (up to 1.45 N). The conducted transparency tests showed the relation between thickness (of the non-woven and fibres) and morphology of the substrate and light transmission. To simulate the in vivo conditions, prior to the light transmission studies, samples were immersed in water. All the samples exhibited high transparency after immersion in water (>80%). The impact of various morphologies was observed in the in vitro studies. All the samples proved high cells viability. Moreover, the substrate morphology had a significant impact on the orientation and arrangement of the fibroblast cytoskeleton. The aligned fibres were oriented in exactly the same direction. The conducted research proved that, by altering the non-wovens microstructure, the properties can be adjusted so as to induce the desirable cellular reaction. This indicates the high potential of electrospun fibres in terms of modulating the corneal cell behaviour in response to the implanted substrate.
Collapse
Affiliation(s)
- Roksana Kurpanik
- Department of Biomaterials and Composites, Faculty of Materials Science and Ceramics, AGH University of Krakow, 30-059 Krakow, Poland.
| | - Marcin Gajek
- Department of Ceramics and Refractories, Faculty of Materials Science and Ceramics, AGH University of Krakow, 30-059 Krakow, Poland
| | - Karol Gryń
- Department of Biomaterials and Composites, Faculty of Materials Science and Ceramics, AGH University of Krakow, 30-059 Krakow, Poland
| | - Piotr Jeleń
- Department of Silicate Chemistry and Macromolecular Compounds, Faculty of Materials Science and Ceramics, AGH University of Krakow, 30-059 Krakow, Poland
| | | | - Ewa Stodolak-Zych
- Department of Biomaterials and Composites, Faculty of Materials Science and Ceramics, AGH University of Krakow, 30-059 Krakow, Poland
| |
Collapse
|
6
|
Desai SU, Srinivasan SS, Kumbar SG, Moss IL. Hydrogel-Based Strategies for Intervertebral Disc Regeneration: Advances, Challenges and Clinical Prospects. Gels 2024; 10:62. [PMID: 38247785 PMCID: PMC10815657 DOI: 10.3390/gels10010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Millions of people worldwide suffer from low back pain and disability associated with intervertebral disc (IVD) degeneration. IVD degeneration is highly correlated with aging, as the nucleus pulposus (NP) dehydrates and the annulus fibrosus (AF) fissures form, which often results in intervertebral disc herniation or disc space collapse and related clinical symptoms. Currently available options for treating intervertebral disc degeneration are symptoms control with therapy modalities, and/or medication, and/or surgical resection of the IVD with or without spinal fusion. As such, there is an urgent clinical demand for more effective disease-modifying treatments for this ubiquitous disorder, rather than the current paradigms focused only on symptom control. Hydrogels are unique biomaterials that have a variety of distinctive qualities, including (but not limited to) biocompatibility, highly adjustable mechanical characteristics, and most importantly, the capacity to absorb and retain water in a manner like that of native human nucleus pulposus tissue. In recent years, various hydrogels have been investigated in vitro and in vivo for the repair of intervertebral discs, some of which are ready for clinical testing. In this review, we summarize the latest findings and developments in the application of hydrogel technology for the repair and regeneration of intervertebral discs.
Collapse
Affiliation(s)
- Shivam U. Desai
- Department of Orthopedic Surgery, Central Michigan University, College of Medicine, Saginaw, MI 48602, USA
| | | | | | - Isaac L. Moss
- Department of Orthopedic Surgery, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
7
|
Liu S, Al-Danakh A, Wang H, Sun Y, Wang L. Advancements in scaffold for treating ligament injuries; in vitro evaluation. Biotechnol J 2024; 19:e2300251. [PMID: 37974555 DOI: 10.1002/biot.202300251] [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: 05/29/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 11/19/2023]
Abstract
Tendon/ligament (T/L) injuries are a worldwide health problem that affects millions of people annually. Due to the characteristics of tendons, the natural rehabilitation of their injuries is a very complex and lengthy process. Surgical treatment of a T/L injury frequently necessitates using autologous or allogeneic grafts or synthetic materials. Nonetheless, these alternatives have limitations in terms of mechanical properties and histocompatibility, and they do not permit the restoration of the original biological function of the tissue, which can negatively impact the patient's quality of life. It is crucial to find biological materials that possess the necessary properties for the successful surgical treatment of tissues and organs. In recent years, the in vitro regeneration of tissues and organs from stem cells has emerged as a promising approach for preparing autologous tissue and organs, and cell culture scaffolds play a critical role in this process. However, the biological traits and serviceability of different materials used for cell culture scaffolds vary significantly, which can impact the properties of the cultured tissues. Therefore, this review aims to analyze the differences in the biological properties and suitability of various materials based on scaffold characteristics such as cell compatibility, degradability, textile technologies, fiber arrangement, pore size, and porosity. This comprehensive analysis provides valuable insights to aid in the selection of appropriate scaffolds for in vitro tissue and organ culture.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Abdullah Al-Danakh
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Haowen Wang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuan Sun
- Liaoning Laboratory of Cancer Genomics and Department of Cell Biology, Dalian Medical University, Dalian, China
| | - Lina Wang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
8
|
Park J, Soh H, Jo S, Weon S, Lee SH, Park JA, Lee MK, Kim TH, Sung IH, Lee JK. Scaffold-induced compression enhances ligamentization potential of decellularized tendon graft reseeded with ACL-derived cells. iScience 2023; 26:108521. [PMID: 38162024 PMCID: PMC10755058 DOI: 10.1016/j.isci.2023.108521] [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: 06/15/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Anterior cruciate ligament (ACL) reconstruction is often performed using a tendon graft. However, the predominant synthesis of fibrotic scar tissue (type III collagen) occurs during the healing process of the tendon graft, resulting in a significantly lower mechanical strength than that of normal ACL tissue. In this study, ACL-derived cells were reseeded to the tendon graft, and scaffold-induced compression was applied to test whether the compressive force results in superior cell survival and integration. Given nanofiber polycaprolactone (PCL) scaffold-induced compression, ACL-derived cells reseeded to a tendon graft demonstrated superior cell survival and integration and resulted in higher gene expression levels of type I collagen compared to non-compressed cell-allograft composites in vitro. Translocation of Yes-associated protein (YAP) into the nucleus was correlated with higher expression of type I collagen in the compression group. These data support the hypothesis of a potential role of mechanotransduction in the ligamentization process.
Collapse
Affiliation(s)
- Jinsung Park
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Hyunsoo Soh
- Department of Orthopaedic Surgery, Hanyang University Hospital, Seoul, Republic of Korea
| | - Sungsin Jo
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Subin Weon
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Seung Hoon Lee
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Jeong-Ah Park
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Myung-Kyu Lee
- Department of Research and Development, Korea Public Tissue Bank, Seongnam-si, Gyeonggi-do, Korea
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Disease, Seoul, Republic of Korea
| | - Il-Hoon Sung
- Department of Orthopaedic Surgery, Hanyang University Hospital, Seoul, Republic of Korea
| | - Jin Kyu Lee
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
- Department of Orthopaedic Surgery, Hanyang University Hospital, Seoul, Republic of Korea
| |
Collapse
|
9
|
Han F, Meng Q, Xie E, Li K, Hu J, Chen Q, Li J, Han F. Engineered biomimetic micro/nano-materials for tissue regeneration. Front Bioeng Biotechnol 2023; 11:1205792. [PMID: 37469449 PMCID: PMC10352664 DOI: 10.3389/fbioe.2023.1205792] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023] Open
Abstract
The incidence of tissue and organ damage caused by various diseases is increasing worldwide. Tissue engineering is a promising strategy of tackling this problem because of its potential to regenerate or replace damaged tissues and organs. The biochemical and biophysical cues of biomaterials can stimulate and induce biological activities such as cell adhesion, proliferation and differentiation, and ultimately achieve tissue repair and regeneration. Micro/nano materials are a special type of biomaterial that can mimic the microstructure of tissues on a microscopic scale due to its precise construction, further providing scaffolds with specific three-dimensional structures to guide the activities of cells. The study and application of biomimetic micro/nano-materials have greatly promoted the development of tissue engineering. This review aims to provide an overview of the different types of micro/nanomaterials, their preparation methods and their application in tissue regeneration.
Collapse
Affiliation(s)
- Feng Han
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Qingchen Meng
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - En Xie
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Kexin Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Jie Hu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Qianglong Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Jiaying Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Fengxuan Han
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
Zhang A, Cheng Z, Chen Y, Shi P, Gan W, Zhang Y. Emerging tissue engineering strategies for annulus fibrosus therapy. Acta Biomater 2023:S1742-7061(23)00337-9. [PMID: 37330029 DOI: 10.1016/j.actbio.2023.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/31/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Low back pain is a major public health concern experienced by 80% of the world's population during their lifetime, which is closely associated with intervertebral disc (IVD) herniation. IVD herniation manifests as the nucleus pulposus (NP) protruding beyond the boundaries of the intervertebral disc due to disruption of the annulus fibrosus (AF). With a deepening understanding of the importance of the AF structure in the pathogenesis of intervertebral disc degeneration, numerous advanced therapeutic strategies for AF based on tissue engineering, cellular regeneration, and gene therapy have emerged. However, there is still no consensus concerning the optimal approach for AF regeneration. In this review, we summarized strategies in the field of AF repair and highlighted ideal cell types and pro-differentiation targeting approaches for AF repair, and discussed the prospects and difficulties of implant systems combining cells and biomaterials to guide future research directions. STATEMENT OF SIGNIFICANCE: Low back pain is a major public health concern experienced by 80% of the world's population during their lifetime, which is closely associated with intervertebral disc (IVD) herniation. However, there is still no consensus concerning the optimal approach for annulus fibrosus (AF) regeneration. In this review, we summarized strategies in the field of AF repair and highlighted ideal cell types and pro-differentiation targeting approaches for AF repair, and discussed the prospects and difficulties of implant systems combining cells and biomaterials to guide future research directions.
Collapse
Affiliation(s)
- Anran Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhangrong Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuhang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengzhi Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weikang Gan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yukun Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
11
|
Chen Z, Xiao L, Hu C, Shen Z, Zhou E, Zhang S, Wang Y. Aligned Lovastatin-loaded Electrospun Nanofibers Regulate Collagen Organization and Reduce Scar Formation. Acta Biomater 2023; 164:240-252. [PMID: 37075962 DOI: 10.1016/j.actbio.2023.04.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Excessive scar formation caused by cutaneous injury leads to pruritus, pain, contracture, dyskinesia, and unpleasant appearance. Functional wound dressings are designed to accelerate wound healing and reduce scar formation. In this study, we fabricated aligned or random polycaprolactone/silk fibroin electrospun nanofiber membranes with or without lovastatin loading, and then evaluated their scar-inhibitory effects on wounds under a specific tension direction. The nanofiber membranes exhibited good controlled-release performance, mechanical properties, hydrophilicity, and biocompatibility. Furthermore, nanofibers' perpendicular placement to the tension direction of the wound most effectively reduced scar formation (the scar area decreased by 66.9%) and promoted skin regeneration in vivo. The mechanism was associated with its aligned nanofibers regulated collagen organization in the early stage of wound healing. Moreover, lovastatin-loaded nanofibers inhibited myofibroblast differentiation and migration. Both tension direction-perpendicular topographical cues and lovastatin synergistically inhibited mechanical transduction and fibrosis progression, further reducing scar formation. In summary, our study may provide an effective scar prevention strategy in which individualized dressings can be designed according to the local mechanical force direction of patients' wounds, and the addition of lovastatin can further inhibit scar formation. STATEMENT OF SIGNIFICANCE: In vivo, cells and collagen are always arranged parallel to the tension direction. However, the aligned topographic cues themselves promote myofibroblast differentiation and exacerbate scar formation. Electrospun nanofibers' perpendicular placement to the tension direction of the wound most effectively reduces scar formation and promotes skin regeneration in vivo. The mechanism is associated with its tension direction-perpendicular nanofibers reregulate collagen organization in the early stage of wound healing. In addition, tension direction-perpendicular topographical cue and lovastatin could inhibit mechanical transduction and fibrosis progression synergistically, further reducing scar formation. This study proves that combining topographical cues of wound dressing and drugs would be a promising therapy for clinical scar management.
Collapse
Affiliation(s)
- Zuhan Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan, 430072, China; Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lingfei Xiao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Chaoyu Hu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan, 430072, China
| | - Zixia Shen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan, 430072, China
| | - Encheng Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan, 430072, China
| | - Shichen Zhang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan, 430072, China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
12
|
Chauhan A, Bhatt AD. A review on design of scaffold for osteoinduction: Toward the unification of independent design variables. Biomech Model Mechanobiol 2023; 22:1-21. [PMID: 36121530 DOI: 10.1007/s10237-022-01635-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022]
Abstract
Biophysical stimulus quantifies the osteoinductivity of the scaffold concerning the mechanoregulatory mathematical models of scaffold-assisted cellular differentiation. Consider a set of independent structural variables ($) that comprises bulk porosity levels ([Formula: see text]) and a set of morphological features of the micro-structure ([Formula: see text]) associated with scaffolds, i.e., [Formula: see text]. The literature suggests that biophysical stimulus ([Formula: see text]) is a function of independent structural variables ($). Limited understanding of the functional correlation between biophysical stimulus and structural features results in the lack of the desired osteoinductivity in a scaffold. Consequently, it limits their broad applicability to assist bone tissue regeneration for treating critical-sized bone fractures. The literature indicates the existence of multi-dimensional independent design variable space as a probable reason for the general lack of osteoinductivity in scaffolds. For instance, known morphological features are the size, shape, orientation, continuity, and connectivity of the porous regions in the scaffold. It implies that the number of independent variables ([Formula: see text]) is more than two, i.e., [Formula: see text], which interact and influence the magnitude of [Formula: see text] in a unified manner. The efficiency of standard engineering design procedures to analyze the correlation between dependent variable ([Formula: see text]) and independent variables ($) in 3D mutually orthogonal Cartesian coordinate system diminishes proportionally with the increase in the number of independent variables ([Formula: see text]) (Deb in Optimization for engineering design-algorithms and examples, PHI Learning Private Limited, New Delhi, 2012). Therefore, there is an immediate need to devise a framework that has the potential to quantify the micro-structural's morphological features in a unified manner to increase the prospects of scaffold-assisted bone tissue regeneration.
Collapse
Affiliation(s)
- Atul Chauhan
- Department of Mechanical Engineering, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India.
| | - Amba D Bhatt
- Department of Mechanical Engineering, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India
| |
Collapse
|
13
|
Core-shell oxygen-releasing fibers for annulus fibrosus repair in the intervertebral disc of rats. Mater Today Bio 2023; 18:100535. [PMID: 36654965 PMCID: PMC9841168 DOI: 10.1016/j.mtbio.2022.100535] [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: 09/12/2022] [Revised: 12/15/2022] [Accepted: 12/26/2022] [Indexed: 01/05/2023] Open
Abstract
The repair of annulus fibrosus (AF) defect after discectomy in the intervertebral disc (IVD) has presented a challenge over the past decade. Hostile microenvironments in the IVD, including, compression and hypoxia, are critical issues that require special attention. Till date, little information is available on potential strategies to cope with the hypoxia dilemma in AF defect sites. In this study, perfluorotributylamine (PFTBA) core-shell fibers were fabricated by coaxial electrospinning to construct oxygen-releasing scaffold for promoting endogenous repair in the AF after discectomy. We demonstrated that PFTBA fibers (10% chitosan, chitosan: PCL, 1:6) could release oxygen for up to 144 h. The oxygen released from PFTBA fibers was found to protect annulus fibrosus stem cells (AFSCs) from hypoxia-induced apoptosis. In addition, the PFTBA fibers were able to promote proliferation, migration and extracellular matrix (ECM) production in AFSCs under hypoxia, highlighting their therapeutic potential in AF defect repair. Subsequent in vivo studies demonstrated that oxygen-supplying fibers were capable of ameliorating disc degeneration after discectomy, which was evidenced by improved disc height and morphological integrity in rats with the oxygen-releasing scaffolds. Further transcriptome analysis indicated that differential expression genes (DEGs) were enriched in "oxygen transport" and "angiogenesis", which likely contributed to their beneficial effect on endogenous AF regeneration. In summary, the oxygen-releasing scaffold provides novel insights into the oxygen regulation by bioactive materials and raises the therapeutic possibility of oxygen supply strategies for defect repair in AF, as well as other aerobic tissues.
Collapse
|
14
|
Li W, Zhou P, Yan B, Qi M, Chen Y, Shang L, Guan J, Zhang L, Mao Y. Disc regeneration by injectable fucoidan-methacrylated dextran hydrogels through mechanical transduction and macrophage immunomodulation. J Tissue Eng 2023; 14:20417314231180050. [PMID: 37427012 PMCID: PMC10328174 DOI: 10.1177/20417314231180050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/19/2023] [Indexed: 07/11/2023] Open
Abstract
Modulating a favorable inflammatory microenvironment that facilitates the recovery of degenerated discs is a key strategy in the treatment of intervertebral disc (IVD) degeneration (IDD). More interestingly, well-mechanized tissue-engineered scaffolds have been proven in recent years to be capable of sensing mechanical transduction to enhance the proliferation and activation of nucleus pulposus cells (NPC) and have demonstrated an increased potential in the treatment and recovery of degenerative discs. Additionally, existing surgical procedures may not be suitable for IDD treatment, warranting the requirement of new regenerative therapies for the restoration of disc structure and function. In this study, a light-sensitive injectable polysaccharide composite hydrogel with excellent mechanical properties was prepared using dextrose methacrylate (DexMA) and fucoidan with inflammation-modulating properties. Through numerous in vivo experiments, it was shown that the co-culture of this composite hydrogel with interleukin-1β-stimulated NPCs was able to promote cell proliferation whilst preventing inflammation. Additionally, activation of the caveolin1-yes-associated protein (CAV1-YAP) mechanotransduction axis promoted extracellular matrix (ECM) metabolism and thus jointly promoted IVD regeneration. After injection into an IDD rat model, the composite hydrogel inhibited the local inflammatory response by inducing macrophage M2 polarization and gradually reducing the ECM degradation. In this study, we propose a fucoidan-DexMA composite hydrogel, which provides an attractive approach for IVD regeneration.
Collapse
Affiliation(s)
- Weifeng Li
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
- Department of Orthopedics, Lixin County
People’s Hospital, Bozhou, China
| | - Pinghui Zhou
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
| | - Bomin Yan
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
| | - Meiyao Qi
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
| | - Yedan Chen
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
| | - Lijun Shang
- School of Life Sciences, Bengbu Medical
College, Bengbu, China
| | - Jianzhong Guan
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
| | - Li Zhang
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
| | - Yingji Mao
- Department of Orthopaedics and
Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical
College, Bengbu, China
- Anhui Province Key Laboratory of Tissue
Transplantation, Bengbu Medical College, Bengbu, China
- School of Life Sciences, Bengbu Medical
College, Bengbu, China
| |
Collapse
|
15
|
RANDHAWA AAYUSHI, DEB DUTTA SAYAN, GANGULY KEYA, V. PATIL TEJAL, LUTHFIKASARI RACHMI, LIM KITAEK. Understanding cell-extracellular matrix interactions for topology-guided tissue regeneration. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
16
|
Qian H, He L, Ye Z, Wei Z, Ao J. Decellularized matrix for repairing intervertebral disc degeneration: Fabrication methods, applications and animal models. Mater Today Bio 2022; 18:100523. [PMID: 36590980 PMCID: PMC9800636 DOI: 10.1016/j.mtbio.2022.100523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Intervertebral disc degeneration (IDD)-induced low back pain significantly influences the quality of life, placing a burden on public health systems worldwide. Currently available therapeutic strategies, such as conservative or operative treatment, cannot effectively restore intervertebral disc (IVD) function. Decellularized matrix (DCM) is a tissue-engineered biomaterial fabricated using physical, chemical, and enzymatic technologies to eliminate cells and antigens. By contrast, the extracellular matrix (ECM), including collagen and glycosaminoglycans, which are well retained, have been extensively studied in IVD regeneration. DCM inherits the native architecture and specific-differentiation induction ability of IVD and has demonstrated effectiveness in IVD regeneration in vitro and in vivo. Moreover, significant improvements have been achieved in the preparation process, mechanistic insights, and application of DCM for IDD repair. Herein, we comprehensively summarize and provide an overview of the roles and applications of DCM for IDD repair based on the existing evidence to shed a novel light on the clinical treatment of IDD.
Collapse
Key Words
- (3D), three-dimensional
- (AF), annular fibers
- (AFSC), AF stem cells
- (APNP), acellular hydrogel descendent from porcine NP
- (DAF-G), decellularized AF hydrogel
- (DAPI), 4,6-diamidino-2-phenylindole
- (DCM), decellularized matrix
- (DET), detergent-enzymatic treatment
- (DWJM), Wharton's jelly matrix
- (ECM), extracellular matrix
- (EVs), extracellular vesicles
- (Exos), exosome
- (IDD), intervertebral disc degeneration
- (IVD), intervertebral disc
- (LBP), Low back pain
- (NP), nucleus pulposus
- (NPCS), NP-based cell delivery system
- (PEGDA/DAFM), polyethylene glycol diacrylate/decellularized AF matrix
- (SD), sodium deoxycholate
- (SDS), sodium dodecyl sulfate
- (SIS), small intestinal submucosa
- (TGF), transforming growth factor
- (bFGF), basic fibroblast growth factor
- (hADSCs), human adipose-derived stem cells
- (hDF), human dermal fibroblast
- (iAF), inner annular fibers
- (oAF), outer annular fibers
- (sGAG), sulfated glycosaminoglycan
- Decellularized matrix
- Intervertebral disc degeneration
- Regenerative medicine
- Tissue engineering
Collapse
Affiliation(s)
- Hu Qian
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Li He
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhimin Ye
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, China
- Corresponding author. Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, 410000, China.
| | - Zairong Wei
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Jun Ao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Corresponding author. Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, 563000, China.
| |
Collapse
|
17
|
Bai M, Zhang Z, Chen H, Liu X, Xie J. Paxillin tunes the relationship between cell-matrix and cell-cell adhesions to regulate stiffness-dependent dentinogenesis. Regen Biomater 2022; 10:rbac100. [PMID: 36683745 PMCID: PMC9847533 DOI: 10.1093/rb/rbac100] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/02/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
Mechanical stiffness is recognized as a key physical factor and directs cell function via a mechanotransduction process, from extracellular physical cues to intracellular signaling cascades that affect transcriptional activity. Cells continually receive mechanical signals from both the surrounding matrix and adjacent cells. However, how mechanical stiffness cue at cell-substrate interfaces coordinates cell-cell junctions in guiding mesenchymal stem cell behaviors is poorly understood. Here, polydimethylsiloxane substrates with different stiffnesses were used to study mechanosensation/transduction mechanisms in controlling odontogenic differentiation of dental papilla cells (DPCs). DPC phenotypes (morphology and differentiation) changed in response to the applied force derived from stiff substrates. Significantly, higher expression of paxillin on stiffer substrates promoted DPC dentinogenesis. Upon treatment with siRNA to knockdown paxillin, N-cadherin increased mainly in the cytomembrane at the area of cell-cell contacts, whereas β-catenin decreased in the nuclei. The result of a double luciferase reporter assay showed that stiffness promoted β-catenin binding to TCF, which could coactivate the target genes associated with odontogenic differentiation, as evidenced by bioinformatics analysis. Finally, we determined that the addition of a β-catenin inhibitor suppressed DPC mineralization in all the stiffness groups. Thus, our results indicated that a mechanotransduction process from cell-substrate interactions to cell-cell adhesions was required for DPC odontogenic differentiation under the stimulation of substrate stiffness. This finding suggests that stem cell fate specification under the stimulus of stiffness at the substrates is based on crosstalk between substrate interactions and adherens junctions, which provides an essential mechanism for cell-based tissue engineering.
Collapse
Affiliation(s)
- Mingru Bai
- Correspondence address. E-mail: (M.B.); (J.X.)
| | - Zhaowei Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Huiyu Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyu Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- Correspondence address. E-mail: (M.B.); (J.X.)
| |
Collapse
|
18
|
Sun H, Wang H, Zhang W, Mao H, Li B. Single-cell RNA sequencing reveals resident progenitor and vascularization-associated cell subpopulations in rat annulus fibrosus. J Orthop Translat 2022; 38:256-267. [PMID: 36568849 PMCID: PMC9758498 DOI: 10.1016/j.jot.2022.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 12/14/2022] Open
Abstract
Background One of the main causes of low back pain is intervertebral disc degeneration (IDD). Annulus fibrosus (AF) is important for the integrity and functions of the intervertebral disc (IVD). However, the resident functional cell components such as progenitors and vascularization-associated cells in AF are yet to be fully identified. Purpose Identification of functional AF cell subpopulations including resident progenitors and vascularization-associated cells. Methods In this study, the single-cell RNA sequencing data of rat IVDs from a public database were analyzed using Seurat for cell clustering, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) for functional analysis, StemID for stem cell identification, Monocle and RNA velocity for pseudotime differentiation trajectory validation, single-cell regulatory network inference and clustering (SCENIC) for gene regulatory network (GRN) analysis, and CellChat for cell-cell interaction analysis. Immunostaining on normal and degenerated rat IVDs, as well as human AF, was used for validations. Results From the data analysis, seven AF cell clusters were identified, including two newly discovered functional clusters, the Grem1 + subpopulation and the Lum + subpopulation. The Grem1 + subpopulation had progenitor characteristics, while the Lum + subpopulation was associated with vascularization during IDD. The GRN analysis showed that Sox9 and Id1 were among the key regulators in the Grem1 + subpopulation, and Nr2f2 and Creb5 could be responsible for the vascularization function in the Lum + subpopulation. Cell-cell interaction analysis revealed highly regulated cellular communications between these cells, and multiple signaling networks including PDGF and MIF signaling pathways were involved in the interactions. Conclusions Our results revealed two new functional AF cell subpopulations, with stemness and vascularization induction potential, respectively. The Translational potential of this article These findings complement our knowledge about IVDs, especially the AF, and in return provide potential cell source and regulation targets for IDD treatment and tissue repair. The existence of the cell subpopulations was also validated in human AF, which strengthen the clinical relevance of the findings.
Collapse
Affiliation(s)
- Heng Sun
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Huan Wang
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Weidong Zhang
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Haijiao Mao
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, China,Corresponding author.
| | - Bin Li
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China,Corresponding author. 178 Ganjiang Rd, Rm 201 Bldg 18, Soochow University (North Campus), Suzhou, Jiangsu, 215007, China.
| |
Collapse
|
19
|
Liu Z, Bian Y, Wu G, Fu C. Application of stem cells combined with biomaterial in the treatment of intervertebral disc degeneration. Front Bioeng Biotechnol 2022; 10:1077028. [PMID: 36507272 PMCID: PMC9732431 DOI: 10.3389/fbioe.2022.1077028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022] Open
Abstract
As the world population is aging, intervertebral disc degeneration (IDD) is becoming a global health issue of increasing concern. A variety of disc degeneration diseases (DDDs) have been proven to be associated with IDD, and these illnesses have significant adverse effects on both individuals and society. The application of stem cells in regenerative medicine, such as blood and circulation, has been demonstrated by numerous studies. Similarly, stem cells have made exciting progress in the treatment of IDD. However, due to complex anatomical structures and functional requirements, traditional stem cell injection makes it difficult to meet people's expectations. With the continuous development of tissue engineering and biomaterials, stem cell combined with biomaterials has far more prospects than before. This review aims to objectively and comprehensively summarize the development of stem cells combined with contemporary biomaterials and the difficulties that need to be overcome.
Collapse
Affiliation(s)
- Zongtai Liu
- Department of Spine Surgery, First Hospital of Jilin University, Changchun, China,Department of Orthopedics, Affiliated Hospital of Beihua University, Jilin, China
| | - Yuya Bian
- Jilin Institute of Scientific and Technical Information, Changchun, China
| | - Guangzhi Wu
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China,*Correspondence: Guangzhi Wu, ; Changfeng Fu,
| | - Changfeng Fu
- Department of Spine Surgery, First Hospital of Jilin University, Changchun, China,*Correspondence: Guangzhi Wu, ; Changfeng Fu,
| |
Collapse
|
20
|
Li Z, Zhang Y, Zhao Y, Gao X, Zhu Z, Mao Y, Qian T. Graded-Three-Dimensional Cell-Encapsulating Hydrogel as a Potential Biologic Scaffold for Disc Tissue Engineering. Tissue Eng Regen Med 2022; 19:1001-1012. [PMID: 35962859 PMCID: PMC9478016 DOI: 10.1007/s13770-022-00480-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Intervertebral disk (IVD) degeneration, which can cause lower back pain, is a major predisposing factor for disability and can be managed through multiple approaches. However, there is no satisfactory strategy currently available to reconstruct and recover the natural properties of IVDs after degeneration. As tissue engineering develops, scaffolds with embedded cell cultures have proved critical for the successful regeneration of IVDs. METHODS In this study, an integrated scaffold for IVD replacement was developed. Through scanning electron microscopy and other mechanical measurements, we characterized the physical properties of different hydrogels. In addition, we simulated the physiological structure of natural IVDs. Nucleus pulposus (NP) cells and annulus fibrosus-derived stem cells (AFSCs) were seeded in gelatin methacrylate (GelMA) hydrogel at different concentrations to evaluate cell viability and matrix expression. RESULTS It was found that different concentrations of GelMA hydrogel can provide a suitable environment for cell survival. However, hydrogels with different mechanical properties influence cell adhesion and extracellular matrix component type I collagen, type II collagen, and aggrecan expression. CONCLUSION This tissue-engineered IVD implant had a similar structure and function as the native IVD, with the inner area mimicking the NP tissue and the outer area mimicking the stratified annulus fibrosus tissue. The new integrated scaffold demonstrated a good simulation of disc structure. The preparation of efficient and regeneration-promoting tissue-engineered scaffolds is an important issue that needs to be explored in the future. It is hoped that this work will provide new ideas and methods for the further construction of functional tissue replacement discs.
Collapse
Affiliation(s)
- Zhixiang Li
- Department of Orthopedics, First Affiliated Hospital, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China
| | - Yiwen Zhang
- Department of Orthopedics, First Affiliated Hospital, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Plastic Surgery Institute of Shantou University Medical College, Shantou, 515063, Guangdong, China
| | - Yupeng Zhao
- Department of Orthopedics, First Affiliated Hospital, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
| | - Xubin Gao
- Department of Orthopedics, First Affiliated Hospital, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
| | - Zhonglian Zhu
- Department of Orthopedics, First Affiliated Hospital, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
| | - Yingji Mao
- Department of Orthopedics, First Affiliated Hospital, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China.
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China.
| | - Taibao Qian
- Department of Orthopedics, First Affiliated Hospital, School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China.
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China.
| |
Collapse
|
21
|
Chu G, Zhang W, Han F, Li K, Liu C, Wei Q, Wang H, Liu Y, Han F, Li B. The role of microenvironment in stem cell-based regeneration of intervertebral disc. Front Bioeng Biotechnol 2022; 10:968862. [PMID: 36017350 PMCID: PMC9395990 DOI: 10.3389/fbioe.2022.968862] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/18/2022] [Indexed: 01/07/2023] Open
Abstract
Regenerative medicine for intervertebral disc (IVD) disease, by utilizing chondrocytes, IVD cells, and stem cells, has progressed to clinical trials in the treatment of back pain, and has been studied in various animal models of disc degeneration in the past decade. Stem cells exist in their natural microenvironment, which provides vital dynamic physical and chemical signals for their survival, proliferation and function. Long-term survival, function and fate of mesenchymal stem cells (MSCs) depend on the microenvironment in which they are transplanted. However, the transplanted MSCs and the endogenous disc cells were influenced by the complicated microenvironment in the degenerating disc with the changes of biochemical and biophysical components. It is important to understand how the MSCs and endogenous disc cells survive and thrive in the harsh microenvironment of the degenerative disc. Furthermore, materials containing stem cells and their natural microenvironment have good clinical effects. However, the implantation of tissue engineering IVD (TE-IVD) cannot provide a complete and dynamic microenvironment for MSCs. IVD graft substitutes may need further improvement to provide the best engineered MSC microenvironment. Additionally, the IVD progenitor cells inside the stem cell niches have been regarded as popular graft cells for IVD regeneration. However, it is still unclear whether actual IVD progenitor cells exist in degenerative spinal conditions. Therefore, the purpose of this review is fourfold: to discuss the presence of endogenous stem cells; to review and summarize the effects of the microenvironment in biological characteristics of MSC, especially those from IVD; to explore the feasibility and prospects of IVD graft substitutes and to elaborate state of the art in the use of MSC transplantation for IVD degeneration in vivo as well as their clinical application.
Collapse
Affiliation(s)
- Genglei Chu
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Weidong Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Feng Han
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Kexin Li
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Chengyuan Liu
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Qiang Wei
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Huan Wang
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yijie Liu
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Fengxuan Han
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
| | - Bin Li
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Suzhou Medical College, Soochow University, Suzhou, China
| |
Collapse
|
22
|
Cho S, Shon MJ, Son B, Eun GS, Yoon TY, Park TH. Tension exerted on cells by magnetic nanoparticles regulates differentiation of human mesenchymal stem cells. BIOMATERIALS ADVANCES 2022; 139:213028. [PMID: 35882121 DOI: 10.1016/j.bioadv.2022.213028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/06/2022] [Accepted: 07/10/2022] [Indexed: 06/15/2023]
Abstract
Cells can 'sense' physical cues in the surrounding microenvironment and 'react' by changing their function. Previous studies have focused on regulating the physical properties of the matrix, such as stiffness and topography, thus changing the tension 'felt' by the cell as a result. In this study, by directly applying a quantified magnetic force to the cell, a correlation between differentiation and tension was shown. The magnetic force, quantified by magnetic tweezers, was applied by incorporating magnetotactic bacteria-isolated magnetic nanoparticles (MNPs) in human mesenchymal stem cells. As the applied tension increased, the expression levels of osteogenic differentiation marker genes and proteins were proportionally upregulated. Additionally, the translocation of YAP and RUNX2, deformation of nucleus, and activation of the MAPK signaling pathway were observed in tension-based osteogenic differentiation. Our findings provide a platform for the quantitative control of tension, a key factor in stem cell differentiation, between cells and the matrix using MNPs. Furthermore, these findings improve the understanding of osteogenic differentiation by mechanotransduction.
Collapse
Affiliation(s)
- Sungwoo Cho
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Min Ju Shon
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Boram Son
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Gee Sung Eun
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Tae-Young Yoon
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
23
|
Importance of Matrix Cues on Intervertebral Disc Development, Degeneration, and Regeneration. Int J Mol Sci 2022; 23:ijms23136915. [PMID: 35805921 PMCID: PMC9266338 DOI: 10.3390/ijms23136915] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 01/25/2023] Open
Abstract
Back pain is one of the leading causes of disability worldwide and is frequently caused by degeneration of the intervertebral discs. The discs’ development, homeostasis, and degeneration are driven by a complex series of biochemical and physical extracellular matrix cues produced by and transmitted to native cells. Thus, understanding the roles of different cues is essential for designing effective cellular and regenerative therapies. Omics technologies have helped identify many new matrix cues; however, comparatively few matrix molecules have thus far been incorporated into tissue engineered models. These include collagen type I and type II, laminins, glycosaminoglycans, and their biomimetic analogues. Modern biofabrication techniques, such as 3D bioprinting, are also enabling the spatial patterning of matrix molecules and growth factors to direct regional effects. These techniques should now be applied to biochemically, physically, and structurally relevant disc models incorporating disc and stem cells to investigate the drivers of healthy cell phenotype and differentiation. Such research will inform the development of efficacious regenerative therapies and improved clinical outcomes.
Collapse
|
24
|
Yu Q, Han F, Yuan Z, Zhu Z, Liu C, Tu Z, Guo Q, Zhao R, Zhang W, Wang H, Mao H, Li B, Zhu C. Fucoidan-loaded nanofibrous scaffolds promote annulus fibrosus repair by ameliorating the inflammatory and oxidative microenvironments in degenerative intervertebral discs. Acta Biomater 2022; 148:73-89. [PMID: 35671874 DOI: 10.1016/j.actbio.2022.05.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/23/2022] [Accepted: 05/31/2022] [Indexed: 11/01/2022]
Abstract
Tissue engineering holds potential in the treatment of intervertebral disc degeneration (IDD). However, implantation of tissue engineered constructs may cause foreign body reaction and aggravate the inflammatory and oxidative microenvironment of the degenerative intervertebral disc (IVD). In order to ameliorate the adverse microenvironment of IDD, in this study, we prepared a biocompatible poly (ether carbonate urethane) urea (PECUU) nanofibrous scaffold loaded with fucoidan, a natural marine bioactive polysaccharide which has great anti-inflammatory and antioxidative functions. Compared with pure PECUU scaffold, the fucoidan-loaded PECUU nanofibrous scaffold (F-PECUU) decreased the gene and protein expression related to inflammation and the oxidative stress in the lipopolysaccharide (LPS) induced annulus fibrosus cells (AFCs) significantly (p<0.05). Especially, gene expression of Ill 6 and Ptgs2 was decreased by more than 50% in F-PECUU with 3.0 wt% fucoidan (HF-PECUU). Moreover, the gene and protein expression related to the degradation of extracellular matrix (ECM) were reduced in a fucoidan concentration-dependent manner significantly, with increased almost 3 times gene expression of Col1a2 and Acan in HF-PECUU. Further, in a 'box' defect model, HF-PECUU decreased the expression of COX-2 and deposited more ECM between scaffold layers when compared with pure PECUU. The disc height and nucleus pulposus hydration of repaired IVD reached up to 75% and 85% of those in the sham group. In addition, F-PECUU helped to maintain an integrate tissue structure with a similar compression modulus to that in sham group. Taken together, the F-PECUU nanofibrous scaffolds showed promising potential to promote AF repair in IDD treatment by ameliorating the harsh degenerative microenvironment. STATEMENT OF SIGNIFICANCE: Annulus fibrosus (AF) tissue engineering holds potential in the treatment of intervertebral disc degeneration (IDD), but is restricted by the inflammatory and oxidative microenvironment of degenerative disc. This study developed a biocompatible polyurethane scaffold (F-PECUU) loaded with fucoidan, a marine bioactive polysaccharide, for ameliorating IDD microenvironment and promoting disc regeneration. F-PECUU alleviated the inflammation and oxidative stress caused by lipopolysaccharide and prevented extracellular matrix (ECM) degradation in AF cells. In vivo, it promoted ECM deposition to maintain the height, water content and mechanical property of disc. This work has shown the potential of marine polysaccharides-containing functional scaffolds in IDD treatment by ameliorating the harsh microenvironment accompanied with disc degeneration.
Collapse
Affiliation(s)
- Qifan Yu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Feng Han
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Zhangqin Yuan
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Zhuang Zhu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Changjiang Liu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Zhengdong Tu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Qianping Guo
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Runze Zhao
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Weidong Zhang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Huan Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315000, China.
| | - Bin Li
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China; Department of Orthopaedic Surgery, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang 315000, China; Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215000, China.
| | - Caihong Zhu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China.
| |
Collapse
|
25
|
Guo X, Wang X, Tang H, Ren Y, Li D, Yi B, Zhang Y. Engineering a Mechanoactive Fibrous Substrate with Enhanced Efficiency in Regulating Stem Cell Tenodifferentiation. ACS APPLIED MATERIALS & INTERFACES 2022; 14:23219-23231. [PMID: 35544769 DOI: 10.1021/acsami.2c04294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Electrospun-aligned fibers in ultrathin fineness have previously demonstrated a limited capacity in driving stem cells to differentiate into tendon-like cells. In view of the tendon's mechanoactive nature, endowing such aligned fibrous structure with mechanoactivity to exert in situ mechanical stimulus by itself, namely, without any forces externally applied, is likely to potentiate its efficiency of tenogenic induction. To test this hypothesis, in this study, a shape-memory-capable poly(l-lactide-co-caprolactone) (PLCL) copolymer was electrospun into aligned fibrous form followed by a "stretching-recovery" shape-programming procedure to impart shape memory capability. Thereafter, in the absence of tenogenic supplements, human adipose-derived stem cells (ADSCs) were cultured on the programmed fibrous substrates for a duration of 7 days, and the effects of constrained recovery resultant stress-stiffening on cell morphology, proliferation, and tenogenic differentiation were examined. The results indicate that the in situ enacted mechanical stimulus due to shape memory effect (SME) did not have adverse influence on cell viability and proliferation, but significantly promoted cellular elongation along the direction of fiber alignment. Moreover, it revealed that tendon-specific protein markers such as tenomodulin (TNMD) and tenascin-C (TNC) and gene expression of scleraxis (SCX), TNMD, TNC, and collagen I (COL I) were significantly upregulated on the mechanoactive fibrous substrate with higher recovery stress compared to the counterparts. Mechanistically, the Rho/ROCK signaling pathway was identified to be involved in the substrate self-actuation-induced enhancement in tenodifferentiation. Together, these results suggest that constrained shape recovery stress may be employed as an innovative loading modality to regulate the stem cell tenodifferentiation by presenting the fibrous substrate with an aligned tendon-like topographical cue and an additional mechanoactivity. This newly demonstrated paradigm in modulating stem cell tenodifferentiation may improve the efficacy of tendon tissue engineering strategy for tendon healing and regeneration.
Collapse
Affiliation(s)
- Xuran Guo
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
| | - Xianliu Wang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
| | - Han Tang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
| | - Yajuan Ren
- Longhua Hospital affiliated to the Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Donghong Li
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
| | - Bingcheng Yi
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital affiliated to the Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yanzhong Zhang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital affiliated to the Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| |
Collapse
|
26
|
Ma J, Li J, Hu S, Wang X, Li M, Xie J, Shi Q, Li B, Lafu S, Chen H. Collagen Modified Anisotropic PLA Scaffold as a base for Peripheral Nerve Regeneration. Macromol Biosci 2022; 22:e2200119. [PMID: 35526091 DOI: 10.1002/mabi.202200119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/28/2022] [Indexed: 11/09/2022]
Abstract
Reconstruction of damaged nerves remains a significant unmet challenge in clinical medicine. Topographical and mechanical stimulations play important roles to repair peripheral nerve injury. The synergistic effects of topography and mechanical rigidity may significantly accelerate nerve regeneration. In this work, we developed a nerve-guiding collagen/polylactic acid (PLA) electrospun scaffold to facilitate peripheral nerve repair. The obtained anisotropic PLA electrospun scaffolds simulated the directional arranged structure of nerve realistically and promoted axonal regeneration after sciatic nerve injury when compared with the isotropic PLA electrospun scaffolds. Moreover, the collagen-modified PLA electrospun scaffolds further provided sufficient mechanical support and favorable microenvironment for axon regeneration. In addition, we observed that collagen-modified PLA electrospun scaffolds facilitated the axon regeneration by regulating YAP molecular pathway. Taken together, we engineered collagen-modified anisotropic PLA electrospun scaffolds may be a potential candidate to combine topography and mechanical rigidity for peripheral nerve regeneration. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jinjin Ma
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Jiaying Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China
| | - Sihan Hu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xingran Wang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Meimei Li
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Jile Xie
- Department of Orthopaedic Surgery, the First Affiliated Hospital, Soochow University, Suzhou, China
| | - Qin Shi
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Bin Li
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China
| | - Saiji Lafu
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,Department of Orthopaedic Surgery, the First Affiliated Hospital, Soochow University, Suzhou, China
| | - Hao Chen
- Affiliated Hospital & Medical College of Yangzhou University, Yangzhou, China
| |
Collapse
|
27
|
Wang L, Zhu T, Kang Y, Zhang J, Du J, Gao H, Chen S, Jiang J, Zhao J. Crimped nanofiber scaffold mimicking tendon-to-bone interface for fatty-infiltrated massive rotator cuff repair. Bioact Mater 2022; 16:149-161. [PMID: 35386329 PMCID: PMC8958472 DOI: 10.1016/j.bioactmat.2022.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Electrospun fibers, with proven ability to promote tissue regeneration, are widely being explored for rotator cuff repairing. However, without post treatment, the microstructure of the electrospun scaffold is vastly different from that of natural extracellular matrix (ECM). Moreover, during mechanical loading, the nanofibers slip that hampers the proliferation and differentiation of migrating stem cells. Here, electrospun nanofiber scaffolds, with crimped nanofibers and welded joints to biomimic the intricate natural microstructure of tendon-to-bone insertion, were prepared using poly(ester-urethane)urea and gelatin via electrospinning and double crosslinking by a multi-bonding network densification strategy. The crimped nanofiber scaffold (CNS) features bionic tensile stress and induces chondrogenic differentiation, laying credible basis for in vivo experimentation. After repairing a rabbit massive rotator cuff tear using a CNS for 3 months, the continuous translational tendon-to-bone interface was fully regenerated, and fatty infiltration was simultaneously inhibited. Instead of micro-CT, μCT was employed to visualize the integrity and intricateness of the three-dimensional microstructure of the CNS-induced-healed tendon-to-bone interface at an ultra-high resolution of less than 1 μm. This study sheds light on the correlation between nanofiber post treatment and massive rotator cuff repair and provides a general strategy for crimped nanofiber preparation and tendon-to-bone interface imaging characterization. Electrospun scaffold mimicking the microstructure of ECM was fabricated. The translational microstructure of tendon-to-bone interface was regenerated. Tendon-to-bone interface was 3D visualized with resolution less than 1 μm. Muscle fatty infiltration was inhibited for massive rotator cuff tear.
Collapse
Affiliation(s)
- Liren Wang
- Department of Sports Medicine, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.,Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| | - Tonghe Zhu
- Department of Sports Medicine, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Yuhao Kang
- Department of Sports Medicine, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.,Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| | - Jianguang Zhang
- Department of Medgen Group Research Laboratory, 18 Qinglan 3 Rd, Shenzhen, 518118, China
| | - Juan Du
- Biofunctional Materials Research Group, College of Chemistry and Chemical Engineering, Multidisciplinary Center for Advanced Materials, Institute of Advanced Studies, Shanghai University of Engineering Science, No. 333 Longteng Rd, Shanghai, 201620, China
| | - Haihan Gao
- Biofunctional Materials Research Group, College of Chemistry and Chemical Engineering, Multidisciplinary Center for Advanced Materials, Institute of Advanced Studies, Shanghai University of Engineering Science, No. 333 Longteng Rd, Shanghai, 201620, China.,Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| | - Sihao Chen
- Biofunctional Materials Research Group, College of Chemistry and Chemical Engineering, Multidisciplinary Center for Advanced Materials, Institute of Advanced Studies, Shanghai University of Engineering Science, No. 333 Longteng Rd, Shanghai, 201620, China
| | - Jia Jiang
- Department of Sports Medicine, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.,Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Jinzhong Zhao
- Department of Sports Medicine, Department of Orthopedics, Shanghai Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.,Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| |
Collapse
|
28
|
Tian KK, Huang SC, Xia XX, Qian ZG. Fibrous Structure and Stiffness of Designer Protein Hydrogels Synergize to Regulate Endothelial Differentiation of Bone Marrow Mesenchymal Stem Cells. Biomacromolecules 2022; 23:1777-1788. [PMID: 35312276 DOI: 10.1021/acs.biomac.2c00032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Matrix stiffness and fibrous structure provided by the native extracellular matrix have been increasingly appreciated as important cues in regulating cell behaviors. Recapitulating these physical cues for cell fate regulation remains a challenge due to the inherent difficulties in making mimetic hydrogels with well-defined compositions, tunable stiffness, and structures. Here, we present two series of fibrous and porous hydrogels with tunable stiffness based on genetically engineered resilin-silk-like and resilin-like protein polymers. Using these hydrogels as substrates, the mechanoresponses of bone marrow mesenchymal stem cells to stiffness and fibrous structure were systematically studied. For both hydrogel series, increasing compression modulus from 8.5 to 14.5 and 23 kPa consistently promoted cell proliferation and differentiation. Nonetheless, the promoting effects were more pronounced on the fibrous gels than their porous counterparts at all three stiffness levels. More interestingly, even the softest fibrous gel (8.5 kPa) allowed the stem cells to exhibit higher endothelial differentiation capability than the toughest porous gel (23 kPa). The predominant role of fibrous structure on the synergistic regulation of endothelial differentiation was further explored. It was found that the stiffness signal activated Yes-associated protein (YAP), the main regulator of endothelial differentiation, via spreading of focal adhesions, whereas fibrous structure reinforced YAP activation by promoting the maturation of focal adhesions and associated F-actin alignment. Therefore, our results shed light on the interplay of physical cues in regulating stem cells and may guide the fabrication of designer proteinaceous matrices toward regenerative medicine.
Collapse
Affiliation(s)
- Kai-Kai Tian
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Sheng-Chen Huang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xiao-Xia Xia
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Zhi-Gang Qian
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| |
Collapse
|
29
|
Nowwarote N, Petit S, Ferre FC, Dingli F, Laigle V, Loew D, Osathanon T, Fournier BPJ. Extracellular Matrix Derived From Dental Pulp Stem Cells Promotes Mineralization. Front Bioeng Biotechnol 2022; 9:740712. [PMID: 35155398 PMCID: PMC8829122 DOI: 10.3389/fbioe.2021.740712] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Extracellular matrix (ECM) plays a pivotal role in many physiological processes. ECM macromolecules and associated factors differ according to tissues, impact cell differentiation, and tissue homeostasis. Dental pulp ECM may differ from other oral tissues and impact mineralization. Thus, the present study aimed to identify the matrisome of ECM proteins derived from human dental pulp stem cells (DPSCs) and its ability to regulate mineralization even in cells which do not respond to assaults by mineralization, the human gingival fibroblasts (GF). Methods: ECM were extracted from DPSCs cultured in normal growth medium supplemented with L-ascorbic acid (N-ECM) or in osteogenic induction medium (OM-ECM). ECM decellularization (dECM) was performed using 0.5% triton X-100 in 20 mM ammonium hydroxide after 21 days. Mass spectrometry and proteomic analysis identified and quantified matrisome proteins. Results: The dECM contained ECM proteins but lacked cellular components and mineralization. Interestingly, collagens (COL6A1, COL6A2, and COL6A3) and elastic fibers (FBN1, FBLN2, FN1, and HSPG2) were significantly represented in N-ECM, while annexins (ANXA1, ANXA4, ANXA5, ANXA6, ANXA7, and ANXA11) were significantly overdetected in OM-ECM. GF were reseeded on N-dECM and OM-dECM and cultured in normal or osteogenic medium. GF were able to attach and proliferate on N-dECM and OM-dECM. Both dECM enhanced mineralization of GF at day 14 compared to tissue culture plate (TCP). In addition, OM-dECM promoted higher mineralization of GF than N-dECM although cultured in growth medium. Conclusions: ECM derived from DPSCs proved to be osteoinductive, and this knowledge supported cell-derived ECM can be further utilized for tissue engineering of mineralized tissues.
Collapse
Affiliation(s)
- Nunthawan Nowwarote
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Molecular Oral Pathophysiology, Université de Paris, Sorbonne Université, Paris, France
- Department of Oral Biology, Dental Faculty Garancière, Université de Paris, Paris, France
| | - Stephane Petit
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Molecular Oral Pathophysiology, Université de Paris, Sorbonne Université, Paris, France
| | - Francois Come Ferre
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Molecular Oral Pathophysiology, Université de Paris, Sorbonne Université, Paris, France
| | - Florent Dingli
- Institut Curie, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, PSL Research University, Paris, France
| | - Victor Laigle
- Institut Curie, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, PSL Research University, Paris, France
| | - Damarys Loew
- Institut Curie, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, PSL Research University, Paris, France
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- *Correspondence: Thanaphum Osathanon, ; Benjamin P. J. Fournier,
| | - Benjamin P. J. Fournier
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Molecular Oral Pathophysiology, Université de Paris, Sorbonne Université, Paris, France
- Department of Oral Biology, Dental Faculty Garancière, Université de Paris, Paris, France
- *Correspondence: Thanaphum Osathanon, ; Benjamin P. J. Fournier,
| |
Collapse
|
30
|
Afzali Z, Matsushita T, Kogure A, Masuda T, Azuma T, Kushiro K, Kasama T, Miyake R, Takai M. Cell Adhesion and Migration on Thickness Gradient Bilayer Polymer Brush Surfaces: Effects of Properties of Polymeric Materials of the Underlayer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:2605-2617. [PMID: 35001615 DOI: 10.1021/acsami.1c21453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In the field of tissue engineering and biomaterials, controlling the surface properties and mechanical properties of scaffold materials is crucial and has attracted much attention. Here, two types of bilayer polymer brushes composed of a hydrophilic underlying layer and a cationic surface layer [made of poly(2-aminoethyl methacrylate)] with a thickness gradient were prepared by surface-initiated atom-transfer radical polymerization. To investigate the influence of the stiffness as a mechanical property of the polymer brush on cell behavior, the underlayer was prepared from either 2-methacryloyloxyethyl phosphorylcholine or oligo(ethylene glycol) methyl ether methacrylate, with the bilayers designated as gradient poly(2-methacryloyloxyethyl phosphorylcholine)-block-poly(2-aminoethyl methacrylate) [grad-pMbA] and gradient poly(oligo[ethylene glycol] methyl ether methacrylate)-block-poly(2-aminoethyl methacrylate) [grad-pEGbA], respectively. Characterization of these surfaces was performed by spectroscopic ellipsometry, X-ray reflectivity, and determination of the zeta potential, static contact angle, and force curve. These diblock copolymer brushes with a thickness gradient helped to distinguish the effects of the mechanical and surface properties of the brushes on cell behavior. The attachment and motility of L929 fibroblasts and epithelial MCF 10A cells on the fabricated brushes were then assessed. L929 cells had a round shape on the thin surface layer of grad-pMbA and spread well on thicker areas. In contrast, MCF 10A cells spread well in areas of any thickness of either grad-pMbA or grad-pEGbA. Single MCF 10A cells migrated randomly on grad-pMbA, whereas grouped cells started to climb up along the thickness gradient of grad-pMbA. In contrast, both single and grouped MCF 10A cells migrated randomly on grad-pEGbA. These thickness gradient diblock copolymer brushes are simple, reproducible, and reasonable platforms that can facilitate practical applications of biomaterials, for example, in tissue engineering and biomaterials.
Collapse
Affiliation(s)
- Zahra Afzali
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Taishi Matsushita
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Akinori Kogure
- Shimadzu Techno-Research, Inc., 380-1 Horiyamashita, Hatano-city, Kanagawa 259-1304, Japan
| | - Tsukuru Masuda
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tomoyuki Azuma
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Keiichiro Kushiro
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Toshihiro Kasama
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Ryo Miyake
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Madoka Takai
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
31
|
Han P, Gomez GA, Duda GN, Ivanovski S, Poh PS. Scaffold geometry modulation of mechanotransduction and its influence on epigenetics. Acta Biomater 2022; 163:259-274. [PMID: 35038587 DOI: 10.1016/j.actbio.2022.01.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 02/03/2023]
Abstract
The dynamics of cell mechanics and epigenetic signatures direct cell behaviour and fate, thus influencing regenerative outcomes. In recent years, the utilisation of 2D geometric (i.e. square, circle, hexagon, triangle or round-shaped) substrates for investigating cell mechanics in response to the extracellular microenvironment have gained increasing interest in regenerative medicine due to their tunable physicochemical properties. In contrast, there is relatively limited knowledge of cell mechanobiology and epigenetics in the context of 3D biomaterial matrices, i.e., hydrogels and scaffolds. Scaffold geometry provides biophysical signals that trigger a nucleus response (regulation of gene expression) and modulates cell behaviour and function. In this review, we explore the potential of additive manufacturing to incorporate multi length-scale geometry features on a scaffold. Then, we discuss how scaffold geometry direct cell and nuclear mechanosensing. We further discuss how cell epigenetics, particularly DNA/histone methylation and histone acetylation, are modulated by scaffold features that lead to specific gene expression and ultimately influence the outcome of tissue regeneration. Overall, we highlight that geometry of different magnitude scales can facilitate the assembly of cells and multicellular tissues into desired functional architectures through the mechanotransduction pathway. Moving forward, the challenge confronting biomedical engineers is the distillation of the vast knowledge to incorporate multiscaled geometrical features that would collectively elicit a favourable tissue regeneration response by harnessing the design flexibility of additive manufacturing. STATEMENT OF SIGNIFICANCE: It is well-established that cells sense and respond to their 2D geometric microenvironment by transmitting extracellular physiochemical forces through the cytoskeleton and biochemical signalling to the nucleus, facilitating epigenetic changes such as DNA methylation, histone acetylation, and microRNA expression. In this context, the current review presents a unique perspective and highlights the importance of 3D architectures (dimensionality and geometries) on cell and nuclear mechanics and epigenetics. Insight into current challenges around the study of mechanobiology and epigenetics utilising additively manufactured 3D scaffold geometries will progress biomaterials research in this space.
Collapse
|
32
|
Wei Q, Wang S, Han F, Wang H, Zhang W, Yu Q, Liu C, Ding L, Wang J, Yu L, Zhu C, Li B, Bl, Cz, Cz, Cz, Qw, Sw, Fh, Hw, Wz, Qy, Cl, Ld, Jw, Ly, Cz, Qw. Cellular modulation by the mechanical cues from biomaterials for tissue engineering. BIOMATERIALS TRANSLATIONAL 2021; 2:323-342. [PMID: 35837415 PMCID: PMC9255801 DOI: 10.12336/biomatertransl.2021.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/13/2021] [Accepted: 07/10/2021] [Indexed: 01/17/2023]
Abstract
Mechanical cues from the extracellular matrix (ECM) microenvironment are known to be significant in modulating the fate of stem cells to guide developmental processes and maintain bodily homeostasis. Tissue engineering has provided a promising approach to the repair or regeneration of damaged tissues. Scaffolds are fundamental in cell-based regenerative therapies. Developing artificial ECM that mimics the mechanical properties of native ECM would greatly help to guide cell functions and thus promote tissue regeneration. In this review, we introduce various mechanical cues provided by the ECM including elasticity, viscoelasticity, topography, and external stimuli, and their effects on cell behaviours. Meanwhile, we discuss the underlying principles and strategies to develop natural or synthetic biomaterials with different mechanical properties for cellular modulation, and explore the mechanism by which the mechanical cues from biomaterials regulate cell function toward tissue regeneration. We also discuss the challenges in multimodal mechanical modulation of cell behaviours and the interplay between mechanical cues and other microenvironmental factors.
Collapse
Affiliation(s)
- Qiang Wei
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Shenghao Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Feng Han
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Huan Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Weidong Zhang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qifan Yu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Changjiang Liu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Luguang Ding
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiayuan Wang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Lili Yu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Caihong Zhu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China,Corresponding authors: Caihong Zhu, ; Bin Li,
| | - Bin Li
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China,College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China,China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang Province, China,Corresponding authors: Caihong Zhu, ; Bin Li,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Li C, Bai Q, Lai Y, Tian J, Li J, Sun X, Zhao Y. Advances and Prospects in Biomaterials for Intervertebral Disk Regeneration. Front Bioeng Biotechnol 2021; 9:766087. [PMID: 34746112 PMCID: PMC8569141 DOI: 10.3389/fbioe.2021.766087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Low-back and neck-shoulder pains caused by intervertebral disk degeneration are highly prevalent among middle-aged and elderly people globally. The main therapy method for intervertebral disk degeneration is surgical intervention, including interbody fusion, disk replacement, and diskectomy. However, the stress changes caused by traditional fusion surgery are prone to degeneration of adjacent segments, while non-fusion surgery has problems, such as ossification of artificial intervertebral disks. To overcome these drawbacks, biomaterials that could endogenously regenerate the intervertebral disk and restore the biomechanical function of the intervertebral disk is imperative. Intervertebral disk is a fibrocartilaginous tissue, primarily comprising nucleus pulposus and annulus fibrosus. Nucleus pulposus (NP) contains high water and proteoglycan, and its main function is absorbing compressive forces and dispersing loads from physical activities to other body parts. Annulus fibrosus (AF) is a multilamellar structure that encloses the NP, comprises water and collagen, and supports compressive and shear stress during complex motion. Therefore, different biomaterials and tissue engineering strategies are required for the functional recovery of NP and AF based on their structures and function. Recently, great progress has been achieved on biomaterials for NP and AF made of functional polymers, such as chitosan, collagen, polylactic acid, and polycaprolactone. However, scaffolds regenerating intervertebral disk remain unexplored. Hence, several tissue engineering strategies based on cell transplantation and growth factors have been extensively researched. In this review, we summarized the functional polymers and tissue engineering strategies of NP and AF to endogenously regenerate degenerative intervertebral disk. The perspective and challenges of tissue engineering strategies using functional polymers, cell transplantation, and growth factor for generating degenerative intervertebral disks were also discussed.
Collapse
Affiliation(s)
- Chunxu Li
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Qiushi Bai
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuxiao Lai
- Centre for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jingjing Tian
- Medical Science Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jiahao Li
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing, China
| | - Yu Zhao
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
34
|
Peredo AP, Gullbrand SE, Smith HE, Mauck RL. Putting the Pieces in Place: Mobilizing Cellular Players to Improve Annulus Fibrosus Repair. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:295-312. [PMID: 32907498 PMCID: PMC10799291 DOI: 10.1089/ten.teb.2020.0196] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The intervertebral disc (IVD) is an integral load-bearing tissue that derives its function from its composite structure and extracellular matrix composition. IVD herniations involve the failure of the annulus fibrosus (AF) and the extrusion of the nucleus pulposus beyond the disc boundary. Disc herniations can impinge the neural elements and cause debilitating pain and loss of function, posing a significant burden on individual patients and society as a whole. Patients with persistent symptoms may require surgery; however, surgical intervention fails to repair the ruptured AF and is associated with the risk for reherniation and further disc degeneration. Given the limitations of AF endogenous repair, many attempts have been made toward the development of effective repair approaches that reestablish IVD function. These methods, however, fail to recapitulate the composition and organization of the native AF, ultimately resulting in inferior tissue mechanics and function over time and high rates of reherniation. Harnessing the cellular function of cells (endogenous or exogenous) at the repair site through the provision of cell-instructive cues could enhance AF tissue regeneration and, ultimately, improve healing outcomes. In this study, we review the diverse approaches that have been developed for AF repair and emphasize the potential for mobilizing the appropriate cellular players at the site of injury to improve AF healing. Impact statement Conventional treatments for intervertebral disc herniation fail to repair the annulus fibrosus (AF), increasing the risk for recurrent herniation. The lack of repair devices in the market has spurred the development of regenerative approaches, yet most of these rely on a scarce endogenous cell population to repair large injuries, resulting in inadequate regeneration. This review identifies current and developing strategies for AF repair and highlights the potential for harnessing cellular function to improve AF regeneration. Ideal cell sources, differentiation strategies, and delivery methods are discussed to guide the design of repair systems that leverage specialized cells to achieve superior outcomes.
Collapse
Affiliation(s)
- Ana P. Peredo
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Sarah E. Gullbrand
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Harvey E. Smith
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Robert L. Mauck
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
35
|
Abstract
Due to the ability to differentiate into variety of cell types, mesenchymal stem cells (MSCs) hold promise as source in cell-based therapy for treating injured tissue and degenerative diseases. The potential use of MSCs to replace or repair damaged tissues may depend on the efficient differentiation protocols to derive specialized cells without any negative side effects. Identification of appropriate cues that support the lineage-specific differentiation of stem cells is critical for tissue healing and cellular therapy. Recently, a number of stimuli have been utilized to direct the differentiation of stem cells. Biochemical stimuli such as small molecule, growth factor and miRNA have been traditionally used to regulate the fate of stem cells. In recent years, many studies have reported that biophysical stimuli including cyclic mechanical strain, fluid shear stress, microgravity, electrical stimulation, matrix stiffness and topography can also be sensed by stem cells through mechanical receptors, thus affecting the stem cell behaviors including their differentiation potential. In this paper, we review all the most recent literature on the application of biochemical and biophysical cues on regulating MSC differentiation. An extensive literature search was done using electronic database (Medline/Pubmed). Although there are still some challenges that need to be taken into consideration before translating these methods into clinics, biochemical and biophysical stimulation appears to be an attractive method to manipulate the lineage commitment of MSCs.
Collapse
|
36
|
Yao X, Wang X, Ding J. Exploration of possible cell chirality using material techniques of surface patterning. Acta Biomater 2021; 126:92-108. [PMID: 33684535 DOI: 10.1016/j.actbio.2021.02.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/10/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Consistent left-right (LR) asymmetry or chirality is critical for embryonic development and function maintenance. While chirality on either molecular or organism level has been well established, that on the cellular level has remained an open question for a long time. Although it remains unclear whether chirality exists universally on the cellular level, valuable efforts have recently been made to explore this fundamental topic pertinent to both cell biology and biomaterial science. The development of material fabrication techniques, surface patterning, in particular, has afforded a unique platform to study cell-material interactions. By using patterning techniques, chirality on the cellular level has been examined for cell clusters and single cells in vitro in well-designed experiments. In this review, we first introduce typical fabrication techniques of surface patterning suitable for cell studies and then summarize the main aspects of preliminary evidence of cell chirality on patterned surfaces to date. We finally indicate the limitations of the studies conducted thus far and describe the perspectives of future research in this challenging field. STATEMENT OF SIGNIFICANCE: While both biomacromolecules and organisms can exhibit chirality, it is not yet conclusive whether a cell has left-right (LR) asymmetry. It is important yet challenging to study and reveal the possible existence of cell chirality. By using the technique of surface patterning, the recent decade has witnessed progress in the exploration of possible cell chirality within cell clusters and single cells. Herein, some important preliminary evidence of cell chirality is collected and analyzed. The open questions and perspectives are also described to promote further investigations of cell chirality in biomaterials.
Collapse
|
37
|
Lian M, Sun B, Han Y, Yu B, Xin W, Xu R, Ni B, Jiang W, Hao Y, Zhang X, Shen Y, Qiao Z, Dai K. A low-temperature-printed hierarchical porous sponge-like scaffold that promotes cell-material interaction and modulates paracrine activity of MSCs for vascularized bone regeneration. Biomaterials 2021; 274:120841. [PMID: 33984633 DOI: 10.1016/j.biomaterials.2021.120841] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/31/2021] [Accepted: 04/16/2021] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) secrete paracrine trophic factors that are beneficial for tissue regeneration. In this study, a sponge-like scaffold with hierarchical and interconnected pores was developed using low-temperature deposition modeling (LDM) printing. Its effects on the cellular behavior, especially on the paracrine secretion patterns of MSCs, were comprehensively investigated. We found that compared with the scaffolds printed via the fused deposition modeling (FDM) technique, the LDM-printed sponges enhanced the adhesion, retention, survival, and ingrowth of MSCs and promoted cell-material interactions. Moreover, the paracrine functions of the cultured MSCs on the LDM-printed sponges were improved, with significant secretion of upregulated immunomodulatory, angiogenic, and osteogenic factors. MSCs on the LDM-printed sponges exert beneficial paracrine effects on multiple regenerative processes, including macrophage polarization, tube formation, and osteogenesis, verifying the enhanced immunomodulatory, angiogenic, and osteogenic potential. Further protein function assays indicated that focal adhesion kinase (FAK), downstream AKT, and yes-associated-protein (YAP) signaling might participate in the required mechanotransductive pathways, through which the hierarchical porous structures stimulated the paracrine effects of MSCs. In a rat distal femoral defect model, the MSC-laden LDM-printed sponges significantly promoted vascularized bone regeneration. The results of the present study demonstrate that the hierarchical porous biomimetic sponges prepared via LDM printing have potential applications in tissue engineering based on their cell-material interaction promotion and MSC paracrine function modulation effects. Furthermore, our findings suggest that the optimization of biomaterial properties to direct the paracrine signaling of MSCs would enhance tissue regeneration.
Collapse
Affiliation(s)
- Meifei Lian
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Binbin Sun
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yu Han
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bin Yu
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Weiwei Xin
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China
| | - Ruida Xu
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China
| | - Bing Ni
- School of Life Science, East China Normal University, Shanghai, 200241, China
| | - Wenbo Jiang
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yongqiang Hao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiuyin Zhang
- Department of Prosthodontics, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yi Shen
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China.
| | - Zhiguang Qiao
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China; Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201112, China.
| | - Kerong Dai
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
38
|
Peng Y, Qing X, Lin H, Huang D, Li J, Tian S, Liu S, Lv X, Ma K, Li R, Rao Z, Bai Y, Chen S, Lei M, Quan D, Shao Z. Decellularized Disc Hydrogels for hBMSCs tissue-specific differentiation and tissue regeneration. Bioact Mater 2021; 6:3541-3556. [PMID: 33842740 PMCID: PMC8022111 DOI: 10.1016/j.bioactmat.2021.03.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue specificity, a key factor in the decellularized tissue matrix (DTM), has shown bioactive functionalities in tuning cell fate-e.g., the differentiation of mesenchymal stem cells. Notably, cell fate is also determined by the living microenvironment, including material composition and spatial characteristics. Herein, two neighboring tissues within intervertebral discs, the nucleus pulposus (NP) and annulus fibrosus (AF), were carefully processed into DTM hydrogels (abbreviated DNP-G and DAF-G, respectively) to determine the tissue-specific effects on stem cell fate, such as specific components and different culturing methods, as well as in vivo regeneration. Distinct differences in their protein compositions were identified by proteomic analysis. Interestingly, the fate of human bone marrow mesenchymal stem cells (hBMSCs) also responds to both culturing methods and composition. Generally, hBMSCs cultured with DNP-G (3D) differentiated into NP-like cells, while hBMSCs cultured with DAF-G (2D) underwent AF-like differentiation, indicating a close correlation with the native microenvironments of NP and AF cells, respectively. Furthermore, we found that the integrin-mediated RhoA/LATS/YAP1 signaling pathway was activated in DAF-G (2D)-induced AF-specific differentiation. Additionally, the activation of YAP1 determined the tendency of NP- or AF-specific differentiation and played opposite regulatory effects. Finally, DNP-G and DAF-G specifically promoted tissue regeneration in NP degeneration and AF defect rat models, respectively. In conclusion, DNP-G and DAF-G can specifically determine the fate of stem cells through the integrin-mediated RhoA/LATS/YAP1 signaling pathway, and this tissue specificity is both compositional and spatial, supporting the utilization of tissue-specific DTM in advanced treatments of intervertebral disc degeneration.
Collapse
Key Words
- 2D, two-dimensional
- 3D, three-dimensional
- AF, annulus fibrosus
- Col I–S, collagen type I solution
- DAF, decellularized annulus fibrosus
- DAF-G, decellularized annulus fibrosus hydrogel
- DAF-S, decellularized annulus fibrosus solution
- DNP, decellularized nucleus pulposus
- DNP-G, decellularized nucleus pulposus hydrogel
- DNP-S, decellularized nucleus pulposus solution
- DTM, decellularized tissue matrix
- Decellularized tissue matrix
- Differentiation
- ECM, extracellular matrix
- FAF, fresh annulus fibrosus
- FNP, fresh nucleus pulposus
- IDD, intervertebral disc degeneration
- Intervertebral disc
- MSC, mesenchymal stem cell
- NP, nucleus pulposus
- Tissue specificity
- YAP1
- YAP1, yes-associated protein 1
Collapse
Affiliation(s)
- Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangcheng Qing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Donghua Huang
- Musculoskeletal Tumor Center, Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jinye Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuo Tian
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sheng Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kaige Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Rui Li
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510127, China
| | - Zilong Rao
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510127, China
| | - Ying Bai
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510127, China
| | - Songfeng Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, China
| | - Ming Lei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Corresponding author.
| | - Daping Quan
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510127, China,School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510127, China,Corresponding author. School of Chemistry, Sun Yat-sen University, Guangzhou, 510127, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Corresponding author.
| |
Collapse
|
39
|
Tao SC, Huang JY, Li ZX, Zhan S, Guo SC. Small extracellular vesicles with LncRNA H19 "overload": YAP Regulation as a Tendon Repair Therapeutic Tactic. iScience 2021; 24:102200. [PMID: 33733065 PMCID: PMC7937563 DOI: 10.1016/j.isci.2021.102200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/10/2021] [Accepted: 02/12/2021] [Indexed: 01/20/2023] Open
Abstract
Functional healing of tendon injuries remains a great challenge. Small extracellular vesicles (sEVs) have received attention as pro-regenerative agents. H19 overexpression could bring tendon regenerative ability, but the mechanism is still not fully elucidated, and reliable method for delivery of long non-coding RNAs (LncRNAs) was demanded. We identified the downstream mechanism of H19, the activation of yes-associated protein (YAP) via the H19-PP1-YAP axis. We established tendon stem/progenitor cells (TSPCs) stably overexpressing H19 with CRISPR-dCas9-based hnRNP A2/B1 activation (H19-CP-TSPCs). H19-OL-sEVs (H19 “overloading” sEVs) could be produced effectively from H19-CP-TSPCs. Only H19-OL-sEVs were able to significantly load large amounts of H19 rather than other competitors, and the potential of H19-OL-sEVs to promote tendon healing was far better than that of other competitors. Our study established a relatively reliable method for enrichment of LncRNAs into sEVs, providing new hints for modularized sEV-based therapies, and modularized sEVs represented a potential strategy for tendon regeneration. H19 overexpression enhances tendon regeneration H19 dephosphorylates and activates YAP hnRNP A2/B1 assists the enrichment of H19 into sEVs H19-OL-sEVs promote tendon regeneration
Collapse
Affiliation(s)
- Shi-Cong Tao
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Ji-Yan Huang
- Department of Stomatology, Shanghai Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 185 Pu'an Road, Shanghai 200021, China
| | - Zi-Xiang Li
- Department of Medicine, Soochou University, Suzhou, Jiangsu 215123, China
| | - Shi Zhan
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Shang-Chun Guo
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
40
|
Zhou P, Wei B, Guan J, Chen Y, Zhu Y, Ye Y, Meng Y, Guan J, Mao Y. Mechanical Stimulation and Diameter of Fiber Scaffolds Affect the Differentiation of Rabbit Annulus Fibrous Stem Cells. Tissue Eng Regen Med 2021; 18:49-60. [PMID: 33145743 PMCID: PMC7862471 DOI: 10.1007/s13770-020-00305-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/12/2020] [Accepted: 09/20/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Degeneration of the annulus fibrosus (AF), an important structure of the intervertebral disc, is one of the main causes of degenerative disc disease. Fabrication of scaffolds replicating the stratified microstructure of the AF is critical for the successful regeneration of AF. METHODS In this study, we cultured rabbit AF-derived stem cells (AFSCs) using fabricated electrospun fibrous poly-L-lactic acid scaffolds with different diameters. We applied cyclic tensile strain (CTS) on the scaffolds to regulate the differentiation of AFSCs into specific cell types that resided at the inner, middle, and outer zones of the AF. RESULTS We found that the morphologies of AFSCs on the smaller-fiber-diameter scaffolds were nearly round, whereas spindle-like cells morphologies were observed on large-diameter scaffolds. CTS enhanced these phenomena and made the cells slender. The expression levels of collagen-I in cells increased as a function of the fiber diameter, whereas collagen-II and aggrecan exhibited opposite trends. Moreover, the application of CTS upregulated the gene expressions of collagen-I, collagen-II, and aggrecan. CONCLUSION Overlaying the scaffolds with different CTS-stimulated cells could eventually lead to engineered AF tissues with hierarchical structures that approximated the native AF tissue. Thus, the proposed methodologies could be potentially applied for AF regeneration.
Collapse
Affiliation(s)
- Pinghui Zhou
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu, 233004, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China
| | - Bangguo Wei
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu, 233004, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China
| | - Jingjing Guan
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu, 233004, China
| | - Yu Chen
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China
- School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
- Department of Plastic Surgery, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Yansong Zhu
- School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China
| | - Yuchen Ye
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu, 233004, China
| | - Yue Meng
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu, 233004, China
| | - Jianzhong Guan
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu, 233004, China.
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China.
| | - Yingji Mao
- Department of Orthopedics, First Affiliated Hospital, Bengbu Medical College, Bengbu, 233004, China.
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, 233030, China.
- School of Life Sciences, Bengbu Medical College, Bengbu, 233030, China.
| |
Collapse
|
41
|
Wang S, Hashemi S, Stratton S, Arinzeh TL. The Effect of Physical Cues of Biomaterial Scaffolds on Stem Cell Behavior. Adv Healthc Mater 2021; 10:e2001244. [PMID: 33274860 DOI: 10.1002/adhm.202001244] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/09/2020] [Indexed: 02/06/2023]
Abstract
Stem cells have been sought as a promising cell source in the tissue engineering field due to their proliferative capacity as well as differentiation potential. Biomaterials have been utilized to facilitate the delivery of stem cells in order to improve their engraftment and long-term viability upon implantation. Biomaterials also have been developed as scaffolds to promote stem cell induced tissue regeneration. This review focuses on the latter where the biomaterial scaffold is designed to provide physical cues to stem cells in order to promote their behavior for tissue formation. Recent work that explores the effect of scaffold physical properties, topography, mechanical properties and electrical properties, is discussed. Although still being elucidated, the biological mechanisms, including cell shape, focal adhesion distribution, and nuclear shape, are presented. This review also discusses emerging areas and challenges in clinical translation.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | - Sharareh Hashemi
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | - Scott Stratton
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | | |
Collapse
|
42
|
Davis S, Roldo M, Blunn G, Tozzi G, Roncada T. Influence of the Mechanical Environment on the Regeneration of Osteochondral Defects. Front Bioeng Biotechnol 2021; 9:603408. [PMID: 33585430 PMCID: PMC7873466 DOI: 10.3389/fbioe.2021.603408] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 01/04/2021] [Indexed: 12/20/2022] Open
Abstract
Articular cartilage is a highly specialised connective tissue of diarthrodial joints which provides a smooth, lubricated surface for joint articulation and plays a crucial role in the transmission of loads. In vivo cartilage is subjected to mechanical stimuli that are essential for cartilage development and the maintenance of a chondrocytic phenotype. Cartilage damage caused by traumatic injuries, ageing, or degradative diseases leads to impaired loading resistance and progressive degeneration of both the articular cartilage and the underlying subchondral bone. Since the tissue has limited self-repairing capacity due its avascular nature, restoration of its mechanical properties is still a major challenge. Tissue engineering techniques have the potential to heal osteochondral defects using a combination of stem cells, growth factors, and biomaterials that could produce a biomechanically functional tissue, representative of native hyaline cartilage. However, current clinical approaches fail to repair full-thickness defects that include the underlying subchondral bone. Moreover, when tested in vivo, current tissue-engineered grafts show limited capacity to regenerate the damaged tissue due to poor integration with host cartilage and the failure to retain structural integrity after insertion, resulting in reduced mechanical function. The aim of this review is to examine the optimal characteristics of osteochondral scaffolds. Additionally, an overview on the latest biomaterials potentially able to replicate the natural mechanical environment of articular cartilage and their role in maintaining mechanical cues to drive chondrogenesis will be detailed, as well as the overall mechanical performance of grafts engineered using different technologies.
Collapse
Affiliation(s)
- Sarah Davis
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Marta Roldo
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Gordon Blunn
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Gianluca Tozzi
- Zeiss Global Centre, School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth, United Kingdom
| | - Tosca Roncada
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
43
|
Ejeian F, Razmjou A, Nasr-Esfahani MH, Mohammad M, Karamali F, Ebrahimi Warkiani M, Asadnia M, Chen V. ZIF-8 Modified Polypropylene Membrane: A Biomimetic Cell Culture Platform with a View to the Improvement of Guided Bone Regeneration. Int J Nanomedicine 2020; 15:10029-10043. [PMID: 33335393 PMCID: PMC7737945 DOI: 10.2147/ijn.s269169] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Despite the significant advances in modeling of biomechanical aspects of cell microenvironment, it remains a major challenge to precisely mimic the physiological condition of the particular cell niche. Here, the metal-organic frameworks (MOFs) have been introduced as a feasible platform for multifactorial control of cell-substrate interaction, given the wide range of physical and mechanical properties of MOF materials and their structural flexibility. RESULTS In situ crystallization of zeolitic imidazolate framework-8 (ZIF-8) on the polydopamine (PDA)-modified membrane significantly raised surface energy, wettability, roughness, and stiffness of the substrate. This modulation led to an almost twofold increment in the primary attachment of dental pulp stem cells (DPSCs) compare to conventional plastic culture dishes. The findings indicate that polypropylene (PP) membrane modified by PDA/ZIF-8 coating effectively supports the growth and proliferation of DPSCs at a substantial rate. Further analysis also displayed the exaggerated multilineage differentiation of DPSCs with amplified level of autocrine cell fate determination signals, like BSP1, BMP2, PPARG, FABP4, ACAN, and COL2A. Notably, osteogenic markers were dramatically overexpressed (more than 100-folds rather than tissue culture plate) in response to biomechanical characteristics of the ZIF-8 layer. CONCLUSION Hence, surface modification of cell culture platforms with MOF nanostructures proposed as a powerful nanomedical approach for selectively guiding stem cells for tissue regeneration. In particular, PP/PDA/ZIF-8 membrane presented ideal characteristics for using as a barrier membrane for guided bone regeneration (GBR) in periodontal tissue engineering.
Collapse
Affiliation(s)
- Fatemeh Ejeian
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan73441-81746, Iran
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Amir Razmjou
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan73441-81746, Iran
- UNESCO Center for Membrane Technology, School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Munirah Mohammad
- UNESCO Center for Membrane Technology, School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Fereshteh Karamali
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Mohsen Asadnia
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Vicki Chen
- School of Chemical Engineering, University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
44
|
Feng Q, Gao H, Wen H, Huang H, Li Q, Liang M, Liu Y, Dong H, Cao X. Engineering the cellular mechanical microenvironment to regulate stem cell chondrogenesis: Insights from a microgel model. Acta Biomater 2020; 113:393-406. [PMID: 32629189 DOI: 10.1016/j.actbio.2020.06.046] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 01/07/2023]
Abstract
Biophysical cues (especially mechanical cues) embedded in cellular microenvironments show a critical impact on stem cell fate. Despite the capability of traditional hydrogels to mimic the feature of extracellular matrix (ECM) and tune their physicochemical properties via diverse approaches, their relatively large size not only induces biased results, but also hinders high-throughput screening and analysis. In this paper, a microgel model is proposed to recapitulate the role of 3D mechanical microenvironment on stem cell behaviors especially chondrogenesis in vitro. The small diameter of microgels brings the high surface area to volume ratio and then the enlarged diffusion area and shortened diffusion distance of soluble molecules, leading to uniform distribution of nutrients and negligible biochemical gradient inside microgels. To construct ECM-like microenvironment with tunable mechanical strength, three gelatin/hyaluronic acid hybrid microgels with low, medium and high crosslinking densities, i.e., Gel-HA(L), Gel-HA(M) and Gel-HA(H), are fabricated in microfluidic devices by Michael addition reaction between thiolated gelatin (Gel-SH) and ethylsulfated hyaluronic acid (HA-VS) with different substitution degrees of vinyl sulfone groups. Our results show that mouse bone marrow mesenchymal stem cell (BMSC) proliferation, distribution and chondrogenesis are all closely dependent on mechanical microenvironments in microgels. Noteworthily, BMSCs show a clear trend of differentiating into hyaline cartilage in Gel-HA(L) and fibrocartilage in Gel-HA(M) and Gel-HA(H). Whole transcriptome RNA sequencing reveals that mechanical microenvironment of microgels affects BMSC differentiation via TGF-β/Smad signaling pathway, Hippo signaling pathway and Integrin/YAP/TAZ signaling pathway. We believe this microgel model provides a new way to further explore the interaction between cells and 3D microenvironment. STATEMENT OF SIGNIFICANCE: In recent years, hydrogels have been frequently used to construct 3D microenvironment for cells. However, their relatively large size not only brings biased experimental results, but also limits high-throughput screening and analysis. Herein we propose a gelatin/hyaluronic acid microgel model to explore the effects of 3D cellular mechanical microenvironment (biophysical cues) on BMSC behaviors especially chondrogenesis, which can minimize the interference of biochemical gradients. Our results reveal that BMSC differentiation into either hyaline cartilage or fibrocartilage can be regulated via tailoring the mechanical properties of microgels. Whole transcriptome RNA sequencing proves that "TGF-β/Smad signaling pathway", "Hippo signaling pathway" and "Integrins/YAP/ TAZ signaling pathway" are activated or inhibited in this process.
Collapse
Affiliation(s)
- Qi Feng
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
| | - Huichang Gao
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
| | - Hongji Wen
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
| | - Hanhao Huang
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
| | - Qingtao Li
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Minhua Liang
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
| | - Yang Liu
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
| | - Hua Dong
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P R China.
| | - Xiaodong Cao
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China; National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P R China; Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510641, China.
| |
Collapse
|
45
|
Ong W, Marinval N, Lin J, Nai MH, Chong YS, Pinese C, Sajikumar S, Lim CT, Ffrench-Constant C, Bechler ME, Chew SY. Biomimicking Fiber Platform with Tunable Stiffness to Study Mechanotransduction Reveals Stiffness Enhances Oligodendrocyte Differentiation but Impedes Myelination through YAP-Dependent Regulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003656. [PMID: 32790058 DOI: 10.1002/smll.202003656] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Indexed: 06/11/2023]
Abstract
A key hallmark of many diseases, especially those in the central nervous system (CNS), is the change in tissue stiffness due to inflammation and scarring. However, how such changes in microenvironment affect the regenerative process remains poorly understood. Here, a biomimicking fiber platform that provides independent variation of fiber structural and intrinsic stiffness is reported. To demonstrate the functionality of these constructs as a mechanotransduction study platform, these substrates are utilized as artificial axons and the effects of axon structural versus intrinsic stiffness on CNS myelination are independently analyzed. While studies have shown that substrate stiffness affects oligodendrocyte differentiation, the effects of mechanical stiffness on the final functional state of oligodendrocyte (i.e., myelination) has not been shown prior to this. Here, it is demonstrated that a stiff mechanical microenvironment impedes oligodendrocyte myelination, independently and distinctively from oligodendrocyte differentiation. Yes-associated protein is identified to be involved in influencing oligodendrocyte myelination through mechanotransduction. The opposing effects on oligodendrocyte differentiation and myelination provide important implications for current work screening for promyelinating drugs, since these efforts have focused mainly on promoting oligodendrocyte differentiation. Thus, the platform may have considerable utility as part of a drug discovery program in identifying molecules that promote both differentiation and myelination.
Collapse
Affiliation(s)
- William Ong
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
- NTU Institute for Health Technologies (Health Tech NTU), Interdisciplinary Disciplinary School, Nanyang Technological University, Singapore, 637533, Singapore
| | - Nicolas Marinval
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - Junquan Lin
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - Mui Hoon Nai
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Yee-Song Chong
- Department of Physiology, National University of Singapore, Singapore, 117593, Singapore
- Life Sciences Institute Neurobiology Programme, Centre for Life Sciences, National University of Singapore, Singapore, 117456, Singapore
| | - Coline Pinese
- Max Mousseron Institute of Biomolecules (IBMM), UMR CNRS 5247, University of Montpellier, ENSCM, Montpellier, F-34093, France
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, Singapore, 117593, Singapore
- Life Sciences Institute Neurobiology Programme, Centre for Life Sciences, National University of Singapore, Singapore, 117456, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 117599, Singapore
| | - Charles Ffrench-Constant
- MRC-Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Marie E Bechler
- MRC-Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| |
Collapse
|
46
|
Biomimetic bone regeneration using angle-ply collagen membrane-supported cell sheets subjected to mechanical conditioning. Acta Biomater 2020; 112:75-86. [PMID: 32505802 DOI: 10.1016/j.actbio.2020.05.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023]
Abstract
Bone injuries are common and new strategies are desired for achieving ideal bone regeneration for bone defect repair. Scaffolds with bone-mimicking characteristics may provide an appropriate microenvironment to promote bone regeneration. Meanwhile, mechanical stimulation effectively regulates a wide range of cellular behaviors such as cell proliferation and differentiation. In this study, biomimetic multi-layer cell-collagen constructs with angle-ply structural feature were prepared by assembling micropatterned collagen membranes on which aligned MC3T3-E1 cells were cultured. The anisotropic microgrooved collagen membranes effectively guided the alignment of cells and promoted the osteogenic differentiation of them. To further promote cell differentiation and extracellular matrix production, the multi-layer cell-collagen constructs were cultured under mechanical conditioning through cyclic stretching. It was found that the constructs with both cell alignment and mechanical conditioning resulted in better osteogenic potential than those with cell alignment or mechanical conditioning alone. Upon implantation into the critical-sized calvarial defects of mice, the constructs with both cell alignment and mechanical conditioning achieved best new bone formation efficacy. Together, findings from this study reveal that synergized use of microstructural and mechanical cues may provide an effective new approach toward bone regeneration. STATEMENT OF SIGNIFICANCE: Biomimicking is an effective strategy to promote bone regeneration for repairing bone defects. Although numerous studies which micro-structurally mimicked native bone using various scaffolds, far less studies have paid attention to the mechanical environment of bone. In this study, angle-ply collagen membrane-supported cell sheets were prepared and pre-conditioned using mechanical loading prior to implantation at bone defects. The constructs with cell alignment and subjected to mechanical conditioning resulted in better osteogenic differentiation of cells in vitro and new bone formation in vivo than those with cell alignment or mechanical conditioning alone. Therefore, recapitulation of both microstructural and mechanical features of native bone may result in a synergistic effect and provides an effective approach toward bone regeneration.
Collapse
|
47
|
Liu Y, Li Z, Li J, Yang S, Zhang Y, Yao B, Song W, Fu X, Huang S. Stiffness-mediated mesenchymal stem cell fate decision in 3D-bioprinted hydrogels. BURNS & TRAUMA 2020; 8:tkaa029. [PMID: 32733974 PMCID: PMC7382973 DOI: 10.1093/burnst/tkaa029] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/10/2020] [Indexed: 02/07/2023]
Abstract
Background Hydrogels with tuneable mechanical properties are an attractive material platform for 3D bioprinting. Thus far, numerous studies have confirmed that the biophysical cues of hydrogels, such as stiffness, are known to have a profound impact on mesenchymal stem cell (MSC) differentiation; however, their differentiation potential within 3D-bioprinted hydrogels is not completely understood. Here, we propose a protocol for the exploration of how the stiffness of alginate-gelatin (Alg-Gel) composite hydrogels (the widely used bioink) affects the differentiation of MSCs in the presence or absence of differentiation inducing factors. Methods Two types of Alg-Gel composite hydrogels (Young's modulus: 50 kPa vs. 225 kPa) were bioprinted independently of porosity. Then, stiffness-induced biases towards adipogenic and osteogenic differentiation of the embedded MSCs were analysed by co-staining with alkaline phosphatase (ALP) and oil red O. The expression of specific markers at the gene level was detected after a 3-day culture. Results Confocal microscopy indicated that all tested hydrogels supported MSC growth and viability during the culture period. Higher expression of adipogenic and osteogenic markers (ALP and lipoprotein lipase (LPL)) in stiffer 3D-bioprinted matrices demonstrated a more significant response of MSCs to stiffer hydrogels with respect to differentiation, which was more robust in differentiation-inducing medium. However, the LPL expression in stiffer 3D-bioprinted constructs was reduced at day 3 regardless of the presence of differentiation-inducing factors. Although MSCs embedded in softer hydrogels to some extent proceeded toward adipogenic and osteogenic lineages within a few days, their differentiation seemed to be slower and more limited. Interestingly, the hydrogel itself (without differentiation-inducing factors) exhibited a slight effect on whether MSCs differentiated towards an adipogenic or an osteogenic fate. Considering that the mechano-regulated protein Yes-associated protein (YAP) is involved in MSC fate decisions, we further found that inhibition of YAP significantly downregulated the expression of ALP and LPL in MSCs in stiffer constructs regardless of the induced growth factors present. Conclusions These results demonstrate that the differentiation of MSCs in 3D-bioprinted matrices is dependent on hydrogel stiffness, which emphasizes the importance of biophysical cues as a determinant of cellular behaviour.
Collapse
Affiliation(s)
- Yufan Liu
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Zhao Li
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Jianjun Li
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Siming Yang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Yijie Zhang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Bin Yao
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Wei Song
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Sha Huang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| |
Collapse
|
48
|
Zhang F, King MW. Biodegradable Polymers as the Pivotal Player in the Design of Tissue Engineering Scaffolds. Adv Healthc Mater 2020; 9:e1901358. [PMID: 32424996 DOI: 10.1002/adhm.201901358] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 04/03/2020] [Indexed: 01/15/2023]
Abstract
Biodegradable polymers play a pivotal role in in situ tissue engineering. Utilizing various technologies, researchers have been able to fabricate 3D tissue engineering scaffolds using biodegradable polymers. They serve as temporary templates, providing physical and biochemical signals to the cells and determining the successful outcome of tissue remodeling. Furthermore, a biodegradable scaffold also presents the fourth dimension for tissue engineering, namely time. The properties of the biodegradable polymer change over time, presenting continuously changing features during the degradation process. These changes become more complicated when different materials are combined together to fabricate a composite or heterogeneous scaffold. This review undertakes a systematic analysis of the basic characteristics of biodegradable polymers and describe recent advances in making composite biodegradable scaffolds for in situ tissue engineering and regenerative medicine. The interaction between implanted biodegradable biomaterials and the in vivo environment are also discussed, including the properties and functional changes of the degradable scaffold, the local effect of degradation on the contiguous tissue and their evaluation using both in vitro and in vivo models.
Collapse
Affiliation(s)
- Fan Zhang
- Wilson College of TextilesNorth Carolina State University Raleigh NC 27606 USA
| | - Martin W. King
- Wilson College of TextilesNorth Carolina State University Raleigh NC 27606 USA
- College of TextilesDonghua University Songjiang District Shanghai 201620 China
| |
Collapse
|
49
|
Yang L, Ge L, van Rijn P. Synergistic Effect of Cell-Derived Extracellular Matrices and Topography on Osteogenesis of Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2020; 12:25591-25603. [PMID: 32423202 PMCID: PMC7291345 DOI: 10.1021/acsami.0c05012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/19/2020] [Indexed: 05/03/2023]
Abstract
Cell-derived matrices (CDMs) are an interesting alternative to conventional sources of extracellular matrices (ECMs) as CDMs mimic the natural ECM composition better and are therefore attractive as a scaffolding material for regulating the functions of stem cells. Previous research on stem cell differentiation has demonstrated that both surface topography and CDMs have a significant influence. However, not much focus has been devoted to elucidating possible synergistic effects of CDMs and topography on osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBM-MSCs). In this study, polydimethylsiloxane (PDMS)-based anisotropic topographies (wrinkles) with various topography dimensions were prepared and subsequently combined with native ECMs produced by human fibroblasts that remained on the surface topography after decellularization. The synergistic effect of CDMs combined with topography on osteogenic differentiation of hBM-MSCs was investigated. The results showed that substrates with specific topography dimensions, coated with aligned CDMs, dramatically enhanced the capacity of osteogenesis as investigated using immunofluorescence staining for identifying osteopontin (OPN) and mineralization. Furthermore, the hBM-MSCs on the substrates decorated with CDMs exhibited a higher percentage of (Yes-associated protein) YAP inside the nucleus, stronger cell contractility, and greater formation of focal adhesions, illustrating that enhanced osteogenesis is partly mediated by cellular tension and mechanotransduction following the YAP pathway. Taken together, our findings highlight the importance of ECMs mediating the osteogenic differentiation of stem cells, and the combination of CDMs and topography will be a powerful approach for material-driven osteogenesis.
Collapse
Affiliation(s)
- Liangliang Yang
- Department
of Biomedical Engineering-FB40, University
of Groningen, University Medical Center Groningen, Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J.
Kolff Institute for Biomedical Engineering and Materials Science-FB41,
Groningen, University of Groningen, University
Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Lu Ge
- Department
of Biomedical Engineering-FB40, University
of Groningen, University Medical Center Groningen, Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J.
Kolff Institute for Biomedical Engineering and Materials Science-FB41,
Groningen, University of Groningen, University
Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Patrick van Rijn
- Department
of Biomedical Engineering-FB40, University
of Groningen, University Medical Center Groningen, Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J.
Kolff Institute for Biomedical Engineering and Materials Science-FB41,
Groningen, University of Groningen, University
Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| |
Collapse
|
50
|
Zhuang Z, Zhang Y, Sun S, Li Q, Chen K, An C, Wang L, van den Beucken JJJP, Wang H. Control of Matrix Stiffness Using Methacrylate-Gelatin Hydrogels for a Macrophage-Mediated Inflammatory Response. ACS Biomater Sci Eng 2020; 6:3091-3102. [PMID: 33463297 DOI: 10.1021/acsbiomaterials.0c00295] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The successful tissue integration of a biomedical material is mainly determined by the inflammatory response after implantation. Macrophage behavior toward implanted materials is pivotal to determine the extent of the inflammatory response. Hydrogels with different properties have been developed for various biomedical applications such as wound dressings or cell-loaded scaffolds. However, there is limited investigation available on the effects of hydrogel mechanical properties on macrophage behavior and the further host inflammatory response. To this end, methacrylate-gelatin (GelMA) hydrogels were selected as a model material to study the effect of hydrogel stiffness (2, 10, and 29 kPa) on macrophage phenotype in vitro and the further host inflammatory response in vivo. Our data showed that macrophages seeded on stiffer surfaces tended to induce macrophages toward a proinflammatory (M1) phenotype with increased macrophage spreading, more defined F-actin and focal adhesion staining, and more proinflammatory cytokine secretion and cluster of differentiation (CD) marker expression compared to those on surfaces with a lower stiffness. When these hydrogels were further subcutaneously implanted in mice to assess their inflammatory response, GelMA hydrogels with a lower stiffness showed more macrophage infiltration but thinner fibrotic capsule formation. The more severe inflammatory response can be attributed to the higher percentage of M1 macrophages induced by GelMA hydrogels with a higher stiffness. Collectively, our data demonstrated that macrophage behavior and the further inflammatory response are mechanically regulated by hydrogel stiffness. The macrophage phenotype rather than the macrophage number predominately determined the inflammatory response after the implantation, which can provide new insights into the future design and application of novel hydrogel-based biomaterials.
Collapse
Affiliation(s)
- Zhumei Zhuang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, 2 Linggong Road, High-Tech District, Dalian 116024, P. R. China
| | - Yang Zhang
- Laboratory of Regenerative Biomaterials, Department of Biomedical Engineering, Health Science Center, Shenzhen University, 3688 Nanhai Avenue, Shenzhen 518037, P. R. China.,Regenerative Biomaterials Group, Department of Dentistry, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen 6525GA, The Netherlands
| | - Shengnan Sun
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, 2 Linggong Road, High-Tech District, Dalian 116024, P. R. China
| | - Qiao Li
- Dalian Medical University, South Lvshun Road, Dalian 116044, P. R. China
| | - Kaiwen Chen
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, 2 Linggong Road, High-Tech District, Dalian 116024, P. R. China
| | - Chuanfeng An
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, 2 Linggong Road, High-Tech District, Dalian 116024, P. R. China
| | - Libin Wang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, 2 Linggong Road, High-Tech District, Dalian 116024, P. R. China
| | - Jeroen J J P van den Beucken
- Regenerative Biomaterials Group, Department of Dentistry, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen 6525GA, The Netherlands
| | - Huanan Wang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, 2 Linggong Road, High-Tech District, Dalian 116024, P. R. China
| |
Collapse
|