1
|
Zhou M, Wang Z, Li M, Chen Q, Zhang S, Wang J. Passivated hydrogel interface: Armor against foreign body response and inflammation in small-diameter vascular grafts. Biomaterials 2025; 317:123010. [PMID: 39724767 DOI: 10.1016/j.biomaterials.2024.123010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/30/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
The development of small-diameter vascular grafts (SDVGs) still faces significant challenges, particularly in overcoming blockages within vessels. A key issue is the foreign-body response (FBR) triggered by the implants, which impairs the integration between grafts and native vessels. In this study, we applied an interfacial infiltration strategy to create a stable, hydrophilic, and passivated hydrogel coating on SDVGs. This coating effectively resisted FBR and improved integration between the grafts and host tissue. We also incorporated anthocyanins, an antioxidant, into the hydrogel network to mitigate oxidative stress and promote endothelialization. The hydrogel coating exhibited excellent stability, retaining its integrity during continuous flushing over 15 days. Anthocyanins were released in response to reactive oxygen species (ROS), reducing inflammation and enhancing vascularization in a mouse subcutaneous implantation model. In a rabbit carotid artery replacement model, the SDVGs exhibited rapid endothelialization, guided vascular remodeling, and inhibited calcification, showing strong potential for clinical application. This study presents a straightforward and effective approach to improve the patency rate, endothelialization, and anti-calcification properties of SDVGs by equipping them with a protective anti-FBR and anti-inflammation hydrogel layer.
Collapse
Affiliation(s)
- Mengxue Zhou
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China; NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zihao Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China; NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Mengyu Li
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China; NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qi Chen
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China; NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Shengmin Zhang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China; NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jianglin Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China; NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
2
|
Zhang T, Cheng Z, Zhang Z, Du L, Li Z, Jiang Z, Zheng Z, Kong D, Zhu M, Li W, Xu B. Biomimetic Microchannel Integrated Silk Fibroin Scaffold for Regeneration of Intervertebral Disc Degeneration. Biomater Res 2025; 29:0203. [PMID: 40438125 PMCID: PMC12117185 DOI: 10.34133/bmr.0203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/22/2025] [Accepted: 04/12/2025] [Indexed: 06/01/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) is the primary cause of low back pain, and patients with severe degeneration usually require lumbar fusion or total disc arthroplasty. Lumbar fusion carries the risk of accelerated degeneration of the adjacent intervertebral disc (IVD), and total disc arthroplasty could reduce the risk. However, the clinical application of artificial IVD whose nondegradable properties make it difficult to restore the biological function of the IVD. Therefore, we intend to fabricate a novel biomimetic microchannel integrated silk fibroin scaffold (BMI-SF scaffold) containing annulus fibrosus with oriented cross-microchannels and nucleus pulposus with interconnected porous structure. The BMI-SF scaffold exhibits controllable microchannels as well as excellent biocompatibility and biodegradability. In vitro and in vivo studies have demonstrated that microchannels can direct cells into the BMI-SF scaffold and enhance neovascularization, supplying adequate nutritional support for tissue regeneration. The IVD replacement model showed that the BMI-SF scaffold has superior regenerative effects, such as restoring IVD height and providing motion segments with dynamic mechanical properties akin to the natural IVD. In this study, the BMI-SF scaffold developed using controlled microchannels provides a new strategy for patients with severe IVDD and has broad clinical application prospects.
Collapse
Affiliation(s)
- Tongxing Zhang
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| | - Zhaojun Cheng
- Department of Spine Surgery,
The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Zhen Zhang
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
- Academy of Medical Engineering and Translational Medicine,
Tianjin University, Tianjin 300072, China
| | - Lilong Du
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| | - Zhenhua Li
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| | - Zhuyan Jiang
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| | - Zhaomin Zheng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Deling Kong
- College of Life Sciences,
Nankai University, Tianjin 300071, China
| | - Meifeng Zhu
- College of Life Sciences,
Nankai University, Tianjin 300071, China
| | - Wen Li
- School of Disaster and Emergency Medicine,
Tianjin University, Tianjin 300072, China
| | - Baoshan Xu
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| |
Collapse
|
3
|
Shahin-Shamsabadi A, Cappuccitti J. Muscle-specific acellular ECM fibers made with anchored cell sheet engineering support regeneration in rat models of volumetric muscle loss. Acta Biomater 2025:S1742-7061(25)00348-4. [PMID: 40399155 DOI: 10.1016/j.actbio.2025.05.024] [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: 01/28/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025]
Abstract
Volumetric muscle loss (VML), a condition affecting millions due to trauma, represents a critical unmet need in regenerative medicine, with no established standard of care. This study introduces a de novo therapeutic strategy using tissue-specific skeletal muscle acellular extracellular matrix (aECM) fibers fabricated using scaffold-free Anchored Cell Sheet Engineering technique. These engineered fibers replicate the native ECM composition and microarchitecture of skeletal muscle, incorporating essential structural and basement membrane proteins. In a rat VML model, aECM demonstrated promising regenerative capacity compared to commercial porcine-derived small intestine submucosa (SIS) ECM. Over an 8-week period, compared to contralateral muscle, aECM preserved muscle volume and weight, regulated inflammatory and fibrotic responses, and promoted vascularization. In contrast, SIS was rapidly degraded by week 4 and associated with fibrosis. Force recovery in aECM was lower at the 8-week time point (77 % compared to 91 % in control), but histological and immunohistochemical analyses revealed newly formed, dispersed muscle fibers exclusively in aECM treatment. Importantly, muscle weight was preserved only when aECM was used, resulting in similar normalized force-to-weight across all groups (87 % in aECM vs. 88 % in SIS). The histological analyses further demonstrated ongoing tissue remodeling, indicative of sustained regeneration, in contrast to the premature fibrotic healing observed in the other groups. An innovative quantitative image analysis workflow enabled assessment of spatial tissue heterogeneity through histology and immunohistochemistry images, setting a new standard for regeneration analysis. These findings establish engineered tissue-specific aECM as a transformative approach for VML treatment, laying the groundwork for translation to clinical applications. STATEMENT OF SIGNIFICANCE: The current study introduces a transformative approach to treating volumetric muscle loss (VML) through the development of tissue-specific acellular extracellular matrix (aECM) fibers engineered using a scaffold-free biofabrication platform uniquely suited for recreation of such aECM components. The engineered fibers represent a significant advancement over current commercial options by recreating native ECM composition and microarchitecture while eliminating complications associated with xenogenic materials. Through comprehensive in vivo evaluation in a rat model, it is demonstrated that these engineered fibers maintain muscle mass and promote controlled tissue regeneration, addressing key limitations of existing treatments. The scaffold-free biofabrication of tissue-specific aECM provides a new paradigm for biomaterial design in regenerative medicine.
Collapse
Affiliation(s)
| | - John Cappuccitti
- Evolved.Bio, 280 Joseph Street, Kitchener, Ontario, N2G4Z5, Canada
| |
Collapse
|
4
|
Afshari R, Roy A, Jain S, Lum K, Huang J, Denton S, Annabi N. One-Pot Synthesis of Antibacterial and Antioxidant Self-Healing Bioadhesives Using Ugi Four-Component Reactions. J Biomed Mater Res B Appl Biomater 2025; 113:e35584. [PMID: 40317897 DOI: 10.1002/jbm.b.35584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 05/07/2025]
Abstract
Bioadhesive materials are extensively utilized as alternatives to surgical sutures and wound dressings. Despite significant advancements in their synthesis, current bioadhesives suffer from inadequate mechanical stability, suboptimal wet tissue adhesion, and a lack of inherent antibacterial and antioxidant properties, while requiring multistep synthesis processes, complicating their production for biomedical applications. To address these limitations, we developed a new bioadhesive, named UgiGel, synthesized through a one-pot Ugi four-component reaction (Ugi-4CR). Our strategy utilized gelatin as the backbone, 4-formylphenylboronic acid (4-FPBA) as an aldehyde source for improved adhesion and antibacterial activity, gallic acid (GA) as a carboxylic acid source for improved antioxidant activity and wound healing, and cyclohexyl isocyanide (CyIso) to induce pseudopeptide structures. The internal crosslinking between GA and 4-FPBA via dynamic boronate ester bond formation, triggered by slight pH changes (7.4-7.8) and temperature elevation (25°C-40°C), resulted in the formation of viscoelastic and self-healing hydrogels with water as the only byproduct without the need for initiator/light activation. UgiGel showed higher adhesion to porcine skin tissue (139.8 ± 8.7 kPa) as compared to commercially available bioadhesives, Evicel (26.3 ± 2.6 kPa) and Coseal (19.3 ± 9.9 kPa). It also demonstrated effective antibacterial properties against both Gram-negative and Gram-positive bacteria, as well as antioxidant activity. Additionally, the in vitro studies using NIH-3T3 cells confirmed the biocompatibility of the UgiGel over 7 days of culture. Moreover, in vivo biocompatibility and biodegradation of UgiGel were confirmed via subcutaneous implantation in rats for up to 28 days. Our results demonstrated that UgiGel outperformed commercially available bioadhesives in terms of adhesion, self-healing, and antibacterial activity, without compromising biocompatibility or physical properties, representing a promising multifunctional bioadhesive for wound sealing and repair.
Collapse
Affiliation(s)
- Ronak Afshari
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Arpita Roy
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Saumya Jain
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Kaimana Lum
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Joyce Huang
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Sam Denton
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
5
|
Yang Z, Yu X, Chen J, Ma W, Hao J, Wu C. Bioactive Scaffolds with Ordered Micro/Nano-Scale Topological Surface for Vascularized Bone Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2500975. [PMID: 40190062 DOI: 10.1002/smll.202500975] [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: 01/23/2025] [Revised: 03/07/2025] [Indexed: 05/27/2025]
Abstract
The ordered topological micro/nanostructures of scaffolds play a pivotal role in regulating bone development, remodeling, and regeneration. Nevertheless, achieving the integration of ordered micro/nanostructures into 3D scaffolds remains a formidable challenge. In this context, a brushing-assembly strategy is developed to construct 3D bioactive scaffolds with highly ordered micro/nanostructures. Such an engineered scaffold exhibits a positive regulatory effect on the behavior and fate of bone resident cells, such as mesenchymal stem cells (MSCs) and human umbilical vein endothelial cells (HUVECs), through mechanical stimulation provided by the ordered micro/nanostructures, while also allowing for the precise spatial distribution of multiple cell types through assembly. In vivo experiments demonstrate that scaffolds with ordered nanostructures possess the potential to accelerate vascularized bone regeneration. Overall, this work proposed a universal strategy for the fabrication of bioactive scaffolds with ordered topological micro/nanostructures, bridging the gap between 3D scaffolds and ordered surface microstructures for tissue engineering.
Collapse
Affiliation(s)
- Zhibo Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
| | - Xiaopeng Yu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
| | - Jiajie Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
| | - Wenping Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Jianxin Hao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| |
Collapse
|
6
|
Chen GH, Sia KC, Liu SW, Kao YC, Yang PC, Ho CH, Huang SC, Lee PY, Liang MZ, Chen L, Huang CC. Implantation of MSC spheroid-derived 3D decellularized ECM enriched with the MSC secretome ameliorates traumatic brain injury and promotes brain repair. Biomaterials 2025; 315:122941. [PMID: 39515193 DOI: 10.1016/j.biomaterials.2024.122941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/14/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Traumatic brain injury (TBI) presents substantial clinical challenges, as existing treatments are unable to reverse damage or effectively promote brain tissue regeneration. Although implantable biomaterials have been proposed to support tissue repair by mitigating the adverse microenvironment in injured brains, many fail to replicate the complex composition and architecture of the native extracellular matrix (ECM), resulting in only limited therapeutic outcomes. This study introduces an innovative approach by developing a mesenchymal stem cell (MSC) spheroid-derived three-dimensional (3D) decellularized ECM (dECM) that is enriched with the MSC-derived matrisome and secretome, offering a promising solution for TBI treatment and brain tissue regeneration. Proteomic and cytokine array analyses revealed that 3D dECM retained a diverse array of MSC spheroid-derived matrisome proteins and secretome components, which are crucial for replicating the complexity of native ECM and the therapeutic capabilities of MSCs. These molecules were found to underlie the observed effects of 3D dECM on immunomodulation, proneuritogenesis, and proangiogenesis in our in vitro functional assays. Implantation of 3D dECM into TBI model mice effectively mitigated postinjury tissue damage and promoted brain repair, as evidenced by a reduced brain lesion volume, decreased cell apoptosis, alleviated neuroinflammation, reduced glial scar formation, and increased of neuroblast recruitment to the lesion site. These outcomes culminated in improved motor function recovery in animals, highlighting the multifaceted therapeutic potential of 3D dECM for TBI. In summary, our study elucidates the transformative potential of MSC spheroid-derived bioactive 3D dECM as an implantable biomaterial for effectively mitigating post-TBI neurological damage, paving the way for its broader therapeutic application.
Collapse
Affiliation(s)
- Grace H Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Kee-Chin Sia
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shao-Wen Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ying-Chi Kao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Pei-Ching Yang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chia-Hsin Ho
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shih-Chen Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Peng-Ying Lee
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Min-Zong Liang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
7
|
Yan R, Chen S, Wang B, Liu C, Chen X. Magnetic field-oriented conductive decellularized extracellular matrix hydrogel synergizes with electrical stimulation to promote spinal cord injury repair and electrophysiological function restoration. BIOMATERIALS ADVANCES 2025; 169:214169. [PMID: 39754869 DOI: 10.1016/j.bioadv.2024.214169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/02/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025]
Abstract
Spinal cord injury (SCI) results in electrophysiological and behavioral dysfunction. Electrical stimulation (ES) is considered to be an effective treatment for mild SCI; however, ES is not applicable to severe SCI due to the disruption of electrical conduction caused by tissue defects. Therefore, the use of conductive materials to fill the defects and restore electrical conduction in the spinal cord is a promising therapeutic strategy. In this study, we used ultrasound to composite conductive reduced graphene oxide (rGO) and magnetic Fe3O4 nanoparticles and encapsulated them into gelatin methacryloyl (GelMA) along with decellularized extracellular matrix (dECM) to form a conductive composite hydrogel, rGO/Fe3O4/dECM@GelMA. The rGO/Fe3O4 complexes were able to orientate themselves in the hydrogel with a magnetic field, conferring an orientated electrical conduction function to the hydrogel. The implantation of this composite hydrogel re-established the electrical conduction in the damaged spinal cord and synergized with ES to promote the regeneration of neurons and myelinated axons at the injury site, resulting in the restoration of electrophysiological function of the spinal cord and motor function of the hind limbs of mice. Our study combines a conductive tissue-engineered scaffold with ES therapy to improve the efficacy of ES in severe spinal cord injuries and promote the restoration of spinal cord function.
Collapse
Affiliation(s)
- Ruijia Yan
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center of Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Shu Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center of Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Bixue Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center of Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center of Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Xi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center of Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China.
| |
Collapse
|
8
|
Wang X, He J, Zhang Q, He J, Wang Q. Constructing a 3D co-culture in vitro synovial tissue model for rheumatoid arthritis research. Mater Today Bio 2025; 31:101492. [PMID: 39968522 PMCID: PMC11834117 DOI: 10.1016/j.mtbio.2025.101492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 11/25/2024] [Accepted: 01/12/2025] [Indexed: 02/20/2025] Open
Abstract
The development and exploration of highly effective drugs for rheumatoid arthritis remains an urgent necessity. However, current disease research models are no longer sufficient to meet the rapid development of high-throughput drug screening. In this study, bacterial cellulose simulating the structure of extracellular matrix was used as a 3D culture platform, and THP-1-derived M1 macrophages, representing the inflammatory component, human umbilical vein endothelial cells (HUVECs), simulating the vascular component, and rheumatoid arthritis fibroblast-like synoviocytes (RA-FLSs), embodying the synovial pathology, were co-cultured to simulate the pathological microenvironment in RA synovial tissues, and synovial organoids were constructed. Under three-dimensional (3D) culture conditions, there was a notable upregulation of fatty acid-binding protein 4 (FABP4) in polarized macrophages, and an enhancement of pathological phenotypes in HUVECs and RA-FLSs, mediated through the PI3K/AKT signaling pathway, including cell proliferation, migration, invasion and vascularization. Compared to planar cultures and 2D co-cultures, 3D synovial organoids not only exhibit a broader range of transcriptomic features characteristic of rheumatoid arthritis but also demonstrate increased drug resistance, likely due to the more complex and physiologically relevant cell-cell and cell-matrix interactions present in 3D environments. This model offers a promising path for personalized treatment, accelerating precision medicine in rheumatology.
Collapse
Affiliation(s)
- Xiaocheng Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, 518036, China
| | - Jiaxin He
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, 518036, China
| | - Qiang Zhang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, 518036, China
| | - Juan He
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, 518036, China
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, 518036, China
| |
Collapse
|
9
|
Ding Y, Huang M, Cai P, Yu X, Cui J, Sun B, Mo X, Lu C, Chen J, Wu J. Inflammation-modulating elastic decellularized extracellular matrix scaffold promotes meniscus regeneration. Acta Biomater 2025; 196:93-108. [PMID: 39988032 DOI: 10.1016/j.actbio.2025.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/31/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Scaffold-guided meniscus repair and regeneration show promise for meniscus injuries. Desirable scaffold properties are key to promoting proper tissue remodeling and effective regeneration. Herein, we report an inflammation-modulating elastic decellularized extracellular matrix (ECM) scaffold and evaluate its biological performance on meniscus repair in a rabbit model. An elastic scaffold of decellularized meniscus ECM (dmECM) was first prepared and functionalized with chitosan (CS) and ibuprofen (IBU) to obtain dmECM/CS-IBU scaffold. Our results show that CS and IBU grafting did not affect the overall properties of the dmECM/CS-IBU scaffold, including porous structure, good mechanical strength and elasticity. It promoted chondrocyte proliferation and preserved chondrogenic properties. In addition, both in vitro and in vivo assessments indicate that the dmECM/CS-IBU scaffold showed good anti-inflammatory properties and promoted pro-healing polarization of macrophages. In a partial rabbit meniscus defect model, the dmECM/CS-IBU scaffold showed promotive effects on in situ meniscus repair and preserved cartilage tissue. Therefore, our study provides a feasible strategy for fabricating scaffolds with tissue-specific bioactivity and inflammation-modulating abilities that synergistically promote meniscus repair and regeneration. STATEMENT OF SIGNIFICANCE: Desirable scaffold properties are key to promoting proper tissue remodeling and effective regeneration of meniscus injuries. Herein, elastic decellularized scaffolds were prepared using natural meniscus and successfully grafted with chitosan and the anti-inflammatory drug ibuprofen (dmECM/CS-IBU). The dmECM/CS-IBU scaffold showed a pro-proliferative and phenotype- preserving effect on chondrocytes. In both in vitro and in vivo models, dmECM/CS-IBU scaffolds exhibited wonderful anti-inflammatory activity. In a meniscus white zone defect model, the dmECM/CS-IBU scaffold demonstrated in situ repair of tissue and protection of cartilage tissue. Therefore, we provides a feasible strategy for fabricating scaffolds with tissue-specific bioactivity and inflammation-modulating abilities that synergistically promote meniscus repair and regeneration.
Collapse
Affiliation(s)
- Yangfan Ding
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Department of Biomedical Engineering, Donghua University, Shanghai 201620, China
| | - Moran Huang
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Pengfei Cai
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Department of Biomedical Engineering, Donghua University, Shanghai 201620, China; G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany
| | - Xiao Yu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Department of Biomedical Engineering, Donghua University, Shanghai 201620, China
| | - Jie Cui
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Department of Biomedical Engineering, Donghua University, Shanghai 201620, China
| | - Binbin Sun
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Department of Biomedical Engineering, Donghua University, Shanghai 201620, China
| | - Xiumei Mo
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Department of Biomedical Engineering, Donghua University, Shanghai 201620, China
| | - Changrui Lu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Department of Biomedical Engineering, Donghua University, Shanghai 201620, China.
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Jinglei Wu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Department of Biomedical Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
10
|
Cai H, Zhou L, Hu Y, Zhou T. Machine Learning-Driven Identification of Exosome-Related Genes in Head and Neck Squamous Cell Carcinoma for Prognostic Evaluation and Drug Response Prediction. Biomedicines 2025; 13:780. [PMID: 40299352 PMCID: PMC12024895 DOI: 10.3390/biomedicines13040780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
Background: This study integrated four Gene Expression Omnibus (GEO) datasets to identify disease-specific feature genes in head and neck squamous cell carcinoma (HNSCC) through differential expression analysis with batch effect correction. Methods: The GeneCards database was used to find genes related to exosomes, and samples were categorized into groups with high and low expression levels based on these feature genes. Functional and pathway enrichment analyses (GO, KEGG, and GSEA) were used to investigate the possible biological mechanisms underlying feature genes. A predictive model was produced by using machine learning algorithms (LASSO regression, SVM, and random forest) to find disease-specific feature genes. Receiver operating characteristic (ROC) curve analysis was used to assess the model's effectiveness. The diagnostic model showed excellent predictive accuracy through external data GSE83519 validation. Results: This analysis highlighted 22 genes with significant differential expression. A predictive model based on five important genes (AGRN, TSPAN6, MMP9, HBA1, and PFN2) was produced by using machine learning algorithms. MMP9 and TSPAN6 showed relatively high predictive performance. Using the ssGSEA algorithm, three key genes (MMP9, AGRN, and PFN2) were identified as strongly linked to immune regulation, immune response suppression, and critical signaling pathways involved in HNSCC progression. Matching HNSCC feature gene expression profiles with DSigDB compound signatures uncovered potential therapeutic targets. Molecular docking simulations identified ligands with high binding affinity and stability, notably C5 and Hoechst 33258, which were prioritized for further validation and potential drug development. Conclusions: This study employs a novel diagnostic model for HNSCC constructed using machine learning technology, which can provide support for the early diagnosis of HNSCC and thus contribute to improving patient treatment plans and clinical management strategies.
Collapse
Affiliation(s)
- Hua Cai
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (H.C.); (L.Z.)
| | - Liuqing Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (H.C.); (L.Z.)
| | - Yao Hu
- Department of Otorhinolaryngology, The Central Hospital of Wuhan, Wuhan 430021, China
| | - Tao Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (H.C.); (L.Z.)
| |
Collapse
|
11
|
Hao H, Chen Y, Yu W, Wang X, Wang C, Zhang P, Ji J. Regulating Cell-Material Interfacial Interactions through Selective Cellular Resistance. J Am Chem Soc 2025; 147:9981-9989. [PMID: 40035633 DOI: 10.1021/jacs.5c01433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Regulating the behavior of different types of cells at the material-tissue interface is pivotal for inducing tissue regeneration. Traditional methods enhance target cell activity using specific ligands such as peptides and antibodies, which have stability issues within biological environments. Herein, we show that selective cell resistance can be realized by fine-tuning the material surface chemistry, achieving strong cell selectivity superior to that of extracellular matrix peptides. A certain degree of adsorption resistance differentially affects the adhesion of various types of cells on material surfaces. Taking this principle into account, a polyethylene glycol (PEG) grafted surface was meticulously fine-tuned to selectively support endothelial cells (ECs) while resisting smooth muscle cell attachment. Mechanistic studies identified that the difference in myosin II expression is crucial for cell selectivity. An EC-selective polymer coating for cardiovascular devices was fabricated to promote rapid surface endothelialization and prevent neointimal hyperplasia in vivo.
Collapse
Affiliation(s)
- Hongye Hao
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310027, P. R. China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining 314400, P. R. China
| | - Yifeng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Weijiang Yu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Xingwang Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Cong Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Peng Zhang
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310027, P. R. China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining 314400, P. R. China
| | - Jian Ji
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310027, P. R. China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
- International Research Center for X Polymers, International Campus, Zhejiang University, Haining 314400, P. R. China
- Transvascular Implantation Devices Research Institute China, Hangzhou 310058, P. R. China
| |
Collapse
|
12
|
Tang R, Ding A, Fu C, Umerori K, Rivera M, Alt DS, Carmean CM, Li L, Eppell SJ, Wynshaw-Boris A, Alsberg E. Three-dimensional tissue platform co-laid with native collagen fibers and cells for phenotypic screening of stem cell interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640824. [PMID: 40093051 PMCID: PMC11908223 DOI: 10.1101/2025.02.28.640824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Phenotypic screening of cell-cell and cell-matrix interactions is critical yet challenging for drug discovery and disease modeling. In this study, a scalable 3D tissue platform was developed by co-laying extracted natural insoluble collagen fibers, mesenchymal stem cells, endothelial cells, and neural progenitor cells for phenotypic screening. Cell growth and interactions were enhanced in the co-laid platform, evident through increased cell proliferation, viability, and vascularization. Dense vascular networks rapidly formed through cell-cell and cell-matrix interactions without adding a traditionally needed growth factor set. Both in vitro and implantation studies confirmed that these blood vessels were of human origin. To evaluate the phenotypic screening of cell-cell and cell-matrix interactions, we propose a phenotype screening prototype for stem cell interactions that utilized multivariate analysis encompassing both cell-cell and cell-matrix interactions and demonstrated its effectiveness to screen vasculature formation and autism spectrum disorder (ASD) models. Using the prototype, we confirmed that collagen crosslinking, ROCK, WNT, and YAP pathways impact vasculogenesis. In addition, ASD donor-derived neural progenitor cells can be distinguished from non-ASD control donor-derived neural progenitor cells.
Collapse
|
13
|
Yang G, Pan H, Wei Y, Yang J, Zhang Z, Chen S, Wan W. Directional Mushroom-Derived Scaffold for Microenvironment Regulation in Infected Bone Defects. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2407730. [PMID: 39961061 DOI: 10.1002/adma.202407730] [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: 05/30/2024] [Revised: 12/11/2024] [Indexed: 03/27/2025]
Abstract
Infected bone defects are a common clinical condition, but conventional treatments often fail to achieve the desired outcomes, including addressing antibiotic resistance and preventing nonunion complications. In the presented study, a functionalized decellularized mushroom stem scaffold is developed composed of its naturally aligned channels, Zn2+/curcumin MOFs, hydroxyapatite minerals, and icariin. In vitro, It is found that functionalized acellular mushroom stem scaffold can control bacterial infections through Zn2+/curcumin MOFs. The naturally aligned channels guide bone mesenchymal stem cells (BMSCs) migration, and the components adsorbed on the acellular substrate further promote the migration of BMSCs. Moreover, these functional components further accelerated the polarization of M2 macrophage and osteogenic differentiation of BMSCs. In vivo, the functionalized decellularized mushroom stem scaffold cleared infected bacteria within 3 days, induced extracellular matrix secretion and alignment, and promoted new bone formation to cover defects within 8 weeks. The functionalized decellularized mushroom stem scaffold provides a promising strategy for treating infectious bone defects.
Collapse
Affiliation(s)
- Ganghua Yang
- Department of Orthopaedic Surgery, Institute of Orthopedics of Jiangxi Province and Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Hao Pan
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Yuxuan Wei
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Jianqiu Yang
- Department of Orthopaedic Surgery, Institute of Orthopedics of Jiangxi Province and Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zihan Zhang
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Shixuan Chen
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Wenbing Wan
- Department of Orthopaedic Surgery, Institute of Orthopedics of Jiangxi Province and Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
14
|
Li W, Zhao Y, Cheng Z, Niu F, Ding J, Bai Y, Li Z, Midgley AC, Zhu M. Fine-tuning of porous microchannelled silk fibroin scaffolds for optimal tissue ingrowth. MATERIALS & DESIGN 2025; 251:113711. [DOI: 10.1016/j.matdes.2025.113711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
|
15
|
Jiang C, Miao T, Xing X, Schilling KJ, Lenhard N, Wang L, McDowell S, Nilsson BL, Wang H, Zhang X. Masquelet Inspired in Vivo Engineered Extracellular Matrix as Functional Periosteum for Bone Defect Repair and Reconstruction. Adv Healthc Mater 2025; 14:e2404975. [PMID: 39840608 PMCID: PMC11913577 DOI: 10.1002/adhm.202404975] [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: 12/10/2024] [Revised: 01/09/2025] [Indexed: 01/23/2025]
Abstract
The Masquelet technique that combines a foreign body reaction (FBR)-induced vascularized tissue membrane with staged bone grafting for reconstruction of segmental bone defect has gained wide attention in Orthopedic surgery. The success of Masquelet hinges on its ability to promote formation of a "periosteum-like" FBR-induced membrane at the bone defect site. Inspired by Masquelet's technique, here a novel approach is devised to create periosteum mimetics from decellularized extracellular matrix (dECM), engineered in vivo through FBR, for reconstruction of segmental bone defects. The approach involved 3D printing of polylactic acid (PLA) template with desired pattern/architecture, followed by subcutaneous implantation of the template to form tissue, and depolymerization and decellularization to generate dECM with interconnected channels. The dECM matrices produces from the same mice (autologous) or from different mice (allogenic) are used as a functional periosteum for repair of structural bone allograft in a murine segmental bone defect model. This study shows that autologous dECM performed better than allogenic dECM, further permitting local delivery of low dose BMP-2 to enhance allograft incorporation. The success of this current approach can establish a new line of versatile, patient-specific, and periosteum-like autologous dECM for bone regeneration, offering personalized therapeutics to patients with impaired healing.
Collapse
Affiliation(s)
- Chen Jiang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Tianfeng Miao
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Xiaojie Xing
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Kevin J Schilling
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Nicholas Lenhard
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Lichen Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Susan McDowell
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Bradley L Nilsson
- Department of Chemistry, University of Rochester, Rochester, NY, 14627, USA
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Xinping Zhang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| |
Collapse
|
16
|
Habiburrohman MR, Jamilludin MA, Cahyati N, Herdianto N, Yusuf Y. Fabrication and in vitro cytocompatibility evaluation of porous bone scaffold based on cuttlefish bone-derived nano-carbonated hydroxyapatite reinforced with polyethylene oxide/chitosan fibrous structure. RSC Adv 2025; 15:5135-5150. [PMID: 39963456 PMCID: PMC11831101 DOI: 10.1039/d4ra08457h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
A novel porous bone scaffold based on nano-carbonated hydroxyapatite reinforced with fibrous-like structured polyethylene oxide/chitosan network (nCHA/PEO/CS) was introduced and fabricated via freeze-drying. Prior to this, the nCHA was synthesized through a hydrothermal reaction based on cuttlefish bone (CFB, Sepia officinalis). The raw cuttlefish bone (raw-CFB) was first decomposed to obtain cuttlefish bone-derived calcium oxide (CaO-CFB) by calcination at 1000 °C, which was used for synthesizing nCHA. The chemical composition analysis showed that the nCHA formed AB-type CHA with a high carbonate content of 7.38 wt%, which is in the range of carbonate content in native bone (2-9 wt%). The Ca/P molar ratio of nCHA was 1.712, very close to the Ca/P of biological apatite of 1.71. Morphological analysis revealed that nCHA consists of nanosized particles, potentially offering a large surface area to volume to promote ion exchange and cell interaction. The excellent physicochemical and morphological properties of nCHA proposed suitability as a bone scaffold precursor combined with PEO and CS. The nCHA/PEO/CS scaffolds were freeze-dried with varying PEO/CS concentrations. Physicochemical analysis indicated that increasing the PEO/CS concentration decreased the crystallinity of the scaffold, causing it to be lower than the nCHA crystallinity, which may be beneficial for cell growth. Morphological analysis revealed that the scaffold structure comprised nCHA cross-linked within a fibrous-like structured PEO/CS network, which appropriately mimics the fibrous structure of extracellular matrix (ECM) in natural bone. However, the nCHA/PEO/CS-11 scaffold formed more appropriate pores with suitable porosity for cell development, blood vessel formation, and nutrient perfusion. The nCHA/PEO/CS-11 scaffold also demonstrated sufficient compressive strength and good swelling behavior, which may favor bone regeneration. The nCHA/PEO/CS-11 scaffold demonstrated high cytocompatibility and facilitated the adherence of MC3T3E1 cells on the scaffold surface. The nCHA/PEO/CS-11 scaffold also promoted cell osteogenic differentiation. Owing to its desirable and suitable characteristics, the nCHA/PEO/CS-11 scaffold is promising in bone tissue engineering.
Collapse
Affiliation(s)
- Musyafa Riziq Habiburrohman
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada Yogyakarta 55281 Indonesia
| | - Muhammad Amir Jamilludin
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada Yogyakarta 55281 Indonesia
| | - Nilam Cahyati
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada Yogyakarta 55281 Indonesia
| | - Nendar Herdianto
- Research Centre for Advanced Material, National Research and Innovation Agency (BRIN) South Tangerang 15314 Indonesia
| | - Yusril Yusuf
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada Yogyakarta 55281 Indonesia
| |
Collapse
|
17
|
Capuani S, Campa‐Carranza JN, Hernandez N, Chua CYX, Grattoni A. Modeling of a Bioengineered Immunomodulating Microenvironment for Cell Therapy. Adv Healthc Mater 2025; 14:e2304003. [PMID: 38215451 PMCID: PMC11239796 DOI: 10.1002/adhm.202304003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Indexed: 01/14/2024]
Abstract
Cell delivery and encapsulation platforms are under development for the treatment of Type 1 Diabetes among other diseases. For effective cell engraftment, these platforms require establishing an immune-protected microenvironment as well as adequate vascularization and oxygen supply to meet the metabolic demands of the therapeutic cells. Current platforms rely on 1) immune isolating barriers and indirect vascularization or 2) direct vascularization with local or systemic delivery of immune modulatory molecules. Supported by experimental data, here a broadly applicable predictive computational model capable of recapitulating both encapsulation strategies is developed. The model is employed to comparatively study the oxygen concentration at different levels of vascularization, transplanted cell density, and spatial distribution, as well as with codelivered adjuvant cells. The model is then validated to be predictive of experimental results of oxygen pressure and local and systemic drug biodistribution in a direct vascularization device with local immunosuppressant delivery. The model highlights that dense vascularization can minimize cell hypoxia while allowing for high cell loading density. In contrast, lower levels of vascularization allow for better drug localization reducing systemic dissemination. Overall, it is shown that this model can serve as a valuable tool for the development and optimization of platform technologies for cell encapsulation.
Collapse
Affiliation(s)
- Simone Capuani
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
- College of Materials Science and Opto‐Electronic TechnologyUniversity of Chinese Academy of Science (UCAS)Beijing100049China
| | - Jocelyn Nikita Campa‐Carranza
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
- School of Medicine and Health SciencesTecnologico de MonterreyMonterreyNL64710Mexico
| | - Nathanael Hernandez
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
| | | | - Alessandro Grattoni
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
- Department of SurgeryHouston Methodist HospitalHoustonTX77030USA
- Department of Radiation OncologyHouston Methodist HospitalHoustonTX77030USA
| |
Collapse
|
18
|
Wang C, Yao H, Shi J, Zhang Z, Cong B, Wu Z, Shang X, Hu X, Yang J, Sun H, Gu Z, Cheng G, Chong H, Wang DA, Zhao Y. Injectable tissue-engineered human cartilage matrix composite fibrin glue for regeneration of articular cartilage defects. BIOMATERIALS ADVANCES 2025; 167:214095. [PMID: 39504587 DOI: 10.1016/j.bioadv.2024.214095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/20/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024]
Abstract
Due to the lack of blood vessels and nerves, the ability of cartilage to repair itself is limited, and the injury of articular cartilage urgently needs effective treatment. Currently, the limitation of clinical repair for cartilage defects is that it is difficult to form pure hyaline cartilage repair, and the source of cartilage tissue and cells is limited. To obtain high-purity regenerated hyaline cartilage, we proposed to construct an injectable hydrogel precursor by using human living hyaline cartilage graft (hLhCG) secreted by human chondrocytes as the dispersed phase and fibrinogen solution as the continuous phase, by double injection with thrombin, three-dimensional network hydrogel structure was formed under the action of thrombin to repair joint defects. The component phenotypes of hLhCG and biomechanical properties of composite gel scaffolds were verified. After 12 weeks of injection of the mixed phase at the defect site, the regenerated tissues are similar in composition to adjacent natural tissues and exhibit similar biomechanical properties. The phenotype of regenerated cartilage was verified, confirming the successful regeneration of hyaline cartilage.
Collapse
Affiliation(s)
- Chirun Wang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China.
| | - Junli Shi
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, 999077, Hong Kong Special Administrative Region
| | - Bo Cong
- Department of Osteoarthropathy, Yantaishan Hospital, Yantai 264001, Shandong, PR China
| | - Zhonglian Wu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Xianfeng Shang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Xu Hu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, 999077, Hong Kong Special Administrative Region
| | - Jian Yang
- Clinical Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, PR China
| | - Haidi Sun
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Zehao Gu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Gong Cheng
- Department of Osteoarthropathy, Yantaishan Hospital, Yantai 264001, Shandong, PR China
| | - Hui Chong
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, Jiangsu, PR China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, 999077, Hong Kong Special Administrative Region.
| | - Yuchi Zhao
- Department of Osteoarthropathy, Yantaishan Hospital, Yantai 264001, Shandong, PR China.
| |
Collapse
|
19
|
Salgado García MG, Díaz NF, García López G, Álvarez Maya I, Hernández Jimenez C, Roman Maldonado Y, Mendoza Aguayo DJ, Díaz Martínez NE. Evaluation methods for decellularized tissues: A focus on human amniotic membrane. J Biosci Bioeng 2025; 139:85-94. [PMID: 39592317 DOI: 10.1016/j.jbiosc.2024.10.009] [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: 08/14/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024]
Abstract
Tissue engineering, a multidisciplinary research field aiming to revolutionize regenerative medicine, relies on scaffolds for optimal cell cultures and organ development. Decellularized tissue extracellular matrices (dECM) scaffolds, particularly from human amniotic membrane (hAM), show promise in clinical applications. This review discusses the significance of scaffolds, emphasizing dECM-based hAM scaffolds, delving into ECM complexities, decellularization processes, and evaluation methods. Raman spectroscopy emerges as a non-destructive tool for evaluating ECM preservation, presenting potential for quantifying ECM components in hAM before and after decellularization. The review explores the role of hAM as a biomaterial, detailing its composition and characteristics and emphasizes the importance of evaluating ultrastructural components and suggests Raman spectroscopy as a valuable technique for this purpose.
Collapse
Affiliation(s)
- Miriam Guadalupe Salgado García
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico; Instituto Tecnológico y de Estudios Superiores de Occidente (ITESO), Tlaquepaque, Mexico
| | | | | | - Ikuri Álvarez Maya
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | | | - Yvonne Roman Maldonado
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico; Instituto Tecnológico y de Estudios Superiores de Occidente (ITESO), Tlaquepaque, Mexico
| | | | - Néstor Emmanuel Díaz Martínez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico.
| |
Collapse
|
20
|
Kim D, Kim SG. Cell Homing Strategies in Regenerative Endodontic Therapy. Cells 2025; 14:201. [PMID: 39936992 PMCID: PMC11817319 DOI: 10.3390/cells14030201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
Cell homing, a process that leverages the body's natural ability to recruit cells and repair damaged tissues, presents a promising alternative to cell transplantation methods. Central to this approach is the recruitment of endogenous stem/progenitor cells-such as those from the apical papilla, bone marrow, and periapical tissues-facilitated by chemotactic biological cues. Moreover, biomaterial scaffolds embedded with signaling molecules create supportive environments, promoting cell migration, adhesion, and differentiation for the regeneration of the pulp-dentin complex. By analyzing in vivo animal studies using cell homing strategies, this review explores how biomolecules and scaffold materials enhance the recruitment of endogenous stem cells to the site of damaged dental pulp tissue, thereby promoting repair and regeneration. It also examines the key principles, recent advancements, and current limitations linked to cell homing-based regenerative endodontic therapy, highlighting the interplay of biomaterials, signaling molecules, and their broader clinical implications.
Collapse
Affiliation(s)
- David Kim
- Center for Dental and Craniofacial Research, Columbia University College of Dental Medicine, New York, NY 10032, USA;
| | - Sahng G. Kim
- Division of Endodontics, Columbia University College of Dental Medicine, New York, NY 10032, USA
| |
Collapse
|
21
|
Li H, Li D, Wang X, Zeng Z, Pahlavan S, Zhang W, Wang X, Wang K. Progress in Biomaterials-Enhanced Vascularization by Modulating Physical Properties. ACS Biomater Sci Eng 2025; 11:33-54. [PMID: 39615049 DOI: 10.1021/acsbiomaterials.4c01106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Sufficient vascular system and adequate blood perfusion is crucial for ensuring nutrient and oxygen supply within biomaterials. Actively exploring the optimal physical properties of biomaterials in various application scenarios has provided clues for enhancing vascularization within materials, leading to improved outcomes in tissue engineering and clinical translation. Here we focus on reviewing the physical properties of biomaterials, including pore structure, surface topography, and stiffness, and their effects on promoting vascularization. This angiogenic capability has the potential to provide better standardized research models and personalized treatment strategies for bone regeneration, wound healing, islet transplantation and cardiac repair.
Collapse
Affiliation(s)
- Hao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Dayan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xue Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Ziyuan Zeng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Wei Zhang
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing 102200, China
| | - Xi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| |
Collapse
|
22
|
Puiggalí-Jou A, Hui I, Baldi L, Frischknecht R, Asadikorayem M, Janiak J, Chansoria P, McCabe MC, Stoddart MJ, Hansen KC, Christman KL, Zenobi-Wong M. Biofabrication of anisotropic articular cartilage based on decellularized extracellular matrix. Biofabrication 2025; 17:015044. [PMID: 39757574 DOI: 10.1088/1758-5090/ad9cc2] [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: 08/05/2024] [Accepted: 12/10/2024] [Indexed: 01/07/2025]
Abstract
Tissue-engineered grafts that mimic articular cartilage show promise for treating cartilage injuries. However, engineering cartilage cell-based therapies to match zonal architecture and biochemical composition remains challenging. Decellularized articular cartilage extracellular matrix (dECM) has gained attention for its chondro-inductive properties, yet dECM-based bioinks have limitations in mechanical stability and printability. This study proposes a rapid light-based bioprinting method using a tyrosine-based crosslinking mechanism, which does not require chemical modifications of dECM and thereby preserves its structure and bioactivity. Combining this resin with Filamented Light (FLight) biofabrication enables the creation of cellular, porous, and anisotropic dECM scaffolds composed of aligned microfilaments. Specifically, we focus on the effects of various biopolymer compositions (i.e. hyaluronic acid, collagen I, and dECM) and inner architecture (i.e. bulk light vs FLight) on immune response and cell morphology, and we investigate their influence on nascent ECM production and long-term tissue maturation. Our findings highlight the importance of FLight scaffolds in directing collagen deposition resembling articular cartilage structure and promoting construct maturation, and they emphasize the superiority of biological-rich dECM over single-component materials for engineering articular cartilage, thereby offering new avenues for the development of effective cartilage tissue engineering strategies.
Collapse
Affiliation(s)
- Anna Puiggalí-Jou
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Isabel Hui
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Lucrezia Baldi
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Rea Frischknecht
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Jakub Janiak
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Parth Chansoria
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Maxwell C McCabe
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, 12801 E 17th Ave., Aurora, CO 80045, United States of America
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz 7270, Switzerland
- Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Albert-Ludwigs-University of Freiburg, 79106 Freiburg, Germany
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, 12801 E 17th Ave., Aurora, CO 80045, United States of America
| | - Karen L Christman
- Shu Chien-Gene Lay Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California at San Diego, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, United States of America
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| |
Collapse
|
23
|
Liang Q, Chen S, Hua S, Jiang W, Zhan J, Pu C, Lin R, He Y, Hou H, Qiu X. Biomimetic Versatile Anisotropic, Electroactive Cellulose Hydrogel Scaffolds Tailored from Fern Stem Serving as Nerve Conduit and Cardiac Patch. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2400002. [PMID: 39629973 PMCID: PMC11789595 DOI: 10.1002/advs.202400002] [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: 01/13/2024] [Revised: 11/06/2024] [Indexed: 01/30/2025]
Abstract
Peripheral nerve injury (PNI) and myocardial infarction (MI) are the two most clinically common soft excitable tissue injuries. Both nerve and cardiac tissues exhibit structural anisotropy and electrophysiological activity, providing a wide range of biophysical cues for cell and tissue repair. However, balancing microstructural anisotropy, electroactivity, and biocompatibility is challenging. To address this issue, Dicranopteris linearis (D. linearis) is proposed as a low-perceived value fern plant. Moreover, to enhance its usefulness, it can be designed into a tubular structure and a lamellar structure to bridge the damaged tissue. Therefore, a robust yet simple top-down approach is proposed to designing and fabricating the desired biomimetic versatile hydrogels orienting from the D. linearis to customize for different soft excitable tissue repair applications. These anisotropic electroactive hydrogels performed well as nerve guidance conduits (NGC) and engineered cardiac patches (ECP) in the repair of PNI and MI, respectively. Two birds, one stone. Accordingly, the biomimetic strategy of D. linearis to NGC and D. linearis to ECP is first proposed, opening a new horizon for constructing tissue engineering using natural sources.
Collapse
Affiliation(s)
- Qinghui Liang
- Department of AnatomyGuangdong Provincial Key Laboratory of Construction and Detection in Tissue EngineeringSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| | - Shuhui Chen
- Department of AnatomyGuangdong Provincial Key Laboratory of Construction and Detection in Tissue EngineeringSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| | - Shaofeng Hua
- Department of AnatomyGuangdong Provincial Key Laboratory of Construction and Detection in Tissue EngineeringSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| | - Weihong Jiang
- Department of AnatomyGuangdong Provincial Key Laboratory of Construction and Detection in Tissue EngineeringSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| | - Jiamian Zhan
- Department of AnatomyGuangdong Provincial Key Laboratory of Construction and Detection in Tissue EngineeringSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| | - Chunyi Pu
- Department of AnatomyGuangdong Provincial Key Laboratory of Construction and Detection in Tissue EngineeringSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| | - Rurong Lin
- Department of AnatomyGuangdong Provincial Key Laboratory of Construction and Detection in Tissue EngineeringSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| | - Yutong He
- Department of AnatomyGuangdong Provincial Key Laboratory of Construction and Detection in Tissue EngineeringSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| | - Honghao Hou
- Department of AnatomyGuangdong Provincial Key Laboratory of Construction and Detection in Tissue EngineeringSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| | - Xiaozhong Qiu
- Department of AnatomyGuangdong Provincial Key Laboratory of Construction and Detection in Tissue EngineeringSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| |
Collapse
|
24
|
Sueters J, van Heiningen R, de Vries R, Guler Z, Huirne J, Smit T. Advances in tissue engineering of peripheral nerve and tissue innervation - a systematic review. J Tissue Eng 2025; 16:20417314251316918. [PMID: 39911939 PMCID: PMC11795627 DOI: 10.1177/20417314251316918] [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: 09/10/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Although various options are available to treat injured organs and peripheral nerves, none is without limitations. Auto- and allografts are the first choice of treatment, but tissue survival or functionality is not guaranteed due to often limited vascular and neural networks. In response, tissue-engineered solutions have been developed, yet clinical translations is rare. In this study, a systematic review was performed on tissue-engineered advancements for peripheral nerves and tissues, to aid future developments in bridging the gap toward the clinic by identifying high-potential solutions and unexplored areas. A systematic search was performed in PubMed, Embase, Web of Science, and Scopus until November 9, 2023. Search terms involved "tissue engineering," "guided," "tissue scaffold," and "tissue graft," together with "innervation" and "reinnervation." Original in vivo or in vitro studies meeting the inclusion criteria (tissue-engineered peripheral nerve/innervation of tissue) and no exclusion criteria (no full text available; written in foreign language; nonoriginal article; tissue-engineering of central nervous system; publication before 2012; insufficient study quality or reproducibility) were assessed. A total of 68 out of 3626 original studies were included. Data extraction was based on disease model, cell origin and host species, biomaterial nature and composition, and external stimuli of biological, chemical or physical origin. Although tissue engineering is still in its infancy, explored innervation strategies of today were highlighted with respect to biomaterials, cell types, and external stimuli. The findings emphasize that natural biomaterials, pre-seeding with autologous cell sources, and solutions for reproductive organs are beneficial for future research. Natural biomaterials possess important cues required for cell-material interaction and closely resemble native tissue in terms of biomechanical, geometrical and chemical composition. Autologous cells induce biomaterial functionalization. As these solutions pose no risk of immunorejection and have demonstrated good outcomes, they are most likely to fulfill the clinical demands.
Collapse
Affiliation(s)
- Jayson Sueters
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
| | - Rowan van Heiningen
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Angiogenesis Laboratory, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
| | - Ralph de Vries
- Medical Library, Vrije Universiteit, Amsterdam, The Netherlands
| | - Zeliha Guler
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Obstetrics and Gynecology, Amsterdam UMC – location AMC, Amsterdam, The Netherlands
| | - Judith Huirne
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
| | - Theo Smit
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Medical Biology, Amsterdam UMC – location AMC, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Cheng Y, Lu Y. Physical stimuli-responsive polymeric patches for healthcare. Bioact Mater 2025; 43:342-375. [PMID: 39399837 PMCID: PMC11470481 DOI: 10.1016/j.bioactmat.2024.08.025] [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: 06/26/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 10/15/2024] Open
Abstract
Many chronic diseases have become severe public health problems with the development of society. A safe and efficient healthcare method is to utilize physical stimulus-responsive polymer patches, which may respond to physical stimuli, including light, electric current, temperature, magnetic field, mechanical force, and ultrasound. Under certain physical stimuli, these patches have been widely used in therapy for diabetes, cancer, wounds, hair loss, obesity, and heart diseases since they could realize controllable treatment and reduce the risks of side effects. This review sketches the design principles of polymer patches, including composition, properties, and performances. Besides, control methods of using different kinds of physical stimuli were introduced. Then, the fabrication methods and characterization of patches were explored. Furthermore, recent applications of these patches in the biomedical field were demonstrated. Finally, we discussed the challenges and prospects for its clinical translation. We anticipate that physical stimulus-responsive polymer patches will open up new avenues for healthcare by acting as a platform with multiple functions.
Collapse
Affiliation(s)
- Yifan Cheng
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
26
|
Sueters J, de Boer L, Groenman F, Huirne JAF, Smit TH, Zaat SAJ. A sterilization method for human decellularized vaginal matrices. Sci Rep 2024; 14:31728. [PMID: 39738284 PMCID: PMC11685901 DOI: 10.1038/s41598-024-82409-4] [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: 08/22/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
Vaginal reconstruction is necessary for various congenital and acquired conditions, including vaginal aplasia, trauma, tumors, and gender incongruency. Current surgical and non-surgical treatments often result in significant complications. Decellularized vaginal matrices (DVMs) from human tissue offer a promising alternative, but require effective sterilization to ensure safety and functionality. This study aimed to develop a sterilization method for decellularized human vaginal wall scaffolds. Based on our previously implemented decellularization technique with minor modifications, we designed and examined three sterilization methods consisting of (i) chemical decellularization, (ii) decellularization with additional peracetic acid/hydrogen peroxide (PAA/H2O2); (iii) decellularization with antibiotic and antimycotic (AAE) based treatment. Sterilization efficacy was evaluated through controlled contamination with common vaginal microbes and sterility testing subsequent to each sterilization method. The extracellular matrix (ECM) structure was assessed via histological staining. Decellularization alone reduced some added bacterial contaminants but did not achieve complete sterilization. PAA/H2O2-sterilization resulted in severe ECM damage, rendering it unsuitable. The AAE-treatment demonstrated effective sterilization without compromising the ECM structure. Combined decellularization and AAE-based treatment forms a viable sterilization method for human vaginal wall tissue, maintaining ECM integrity and achieving effective micro-organism elimination. This method holds potential for clinical application in vaginal transplantation.
Collapse
Affiliation(s)
- Jayson Sueters
- Department of Gynaecology, Amsterdam UMC - Location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Leonie de Boer
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC - Location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Freek Groenman
- Department of Obstetrics and Gynecology, Amsterdam Reproduction and Development, Amsterdam UMC - Location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Centre of Expertise on Gender Dysphoria, Amsterdam UMC - Location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Judith A F Huirne
- Department of Gynaecology, Amsterdam UMC - Location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Theo H Smit
- Department of Gynaecology, Amsterdam UMC - Location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam UMC - Location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Sebastian A J Zaat
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC - Location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Yousefi F, Foster LA, Selim OA, Zhao C. Integrating Physical and Biochemical Cues for Muscle Engineering: Scaffolds and Graft Durability. Bioengineering (Basel) 2024; 11:1245. [PMID: 39768063 PMCID: PMC11673930 DOI: 10.3390/bioengineering11121245] [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: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for skeletal muscle regeneration, influenced by a complex interplay of mechanical, biochemical, and molecular cues. Properties of the extracellular matrix (ECM) such as stiffness and alignment guide stem cell fate through mechanosensitive pathways, where forces like shear stress translate into biochemical signals, affecting cell behavior. Aging introduces senescence which disrupts the MuSC niche, leading to reduced regenerative capacity via epigenetic alterations and metabolic shifts. Transplantation further challenges MuSC viability, often resulting in fibrosis driven by dysregulated fibro-adipogenic progenitors (FAPs). Addressing these issues, scaffold designs integrated with pharmacotherapy emulate ECM environments, providing cues that enhance graft functionality and endurance. These scaffolds facilitate the synergy between mechanotransduction and intracellular signaling, optimizing MuSC proliferation and differentiation. Innovations utilizing human pluripotent stem cell-derived myogenic progenitors and exosome-mediated delivery exploit bioactive properties for targeted repair. Additionally, 3D-printed and electrospun scaffolds with adjustable biomechanical traits tackle scalability in treating volumetric muscle loss. Advanced techniques like single-cell RNA sequencing and high-resolution imaging unravel muscle repair mechanisms, offering precise mapping of cellular interactions. Collectively, this interdisciplinary approach fortifies tissue graft durability and MuSC maintenance, propelling therapeutic strategies for muscle injuries and degenerative diseases.
Collapse
Affiliation(s)
- Farbod Yousefi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Lauren Ann Foster
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
- Atlanta Veterans Affairs Medical Center, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Omar A. Selim
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| |
Collapse
|
28
|
Sharma Y, Ghatak S, Sen CK, Mohanty S. Emerging technologies in regenerative medicine: The future of wound care and therapy. J Mol Med (Berl) 2024; 102:1425-1450. [PMID: 39358606 DOI: 10.1007/s00109-024-02493-x] [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: 03/08/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Wound healing, an intricate biological process, comprises orderly phases of simple biological processed including hemostasis, inflammation, angiogenesis, cell proliferation, and ECM remodeling. The regulation of the shift in these phases can be influenced by systemic or environmental conditions. Any untimely transitions between these phases can lead to chronic wounds and scarring, imposing a significant socio-economic burden on patients. Current treatment modalities are largely supportive in nature and primarily involve the prevention of infection and controlling inflammation. This often results in delayed healing and wound complications. Recent strides in regenerative medicine and tissue engineering offer innovative and patient-specific solutions. Mesenchymal stem cells (MSCs) and their secretome have gained specific prominence in this regard. Additionally, technologies like tissue nano-transfection enable in situ gene editing, a need-specific approach without the requirement of complex laboratory procedures. Innovating approaches like 3D bioprinting and ECM bioscaffolds also hold the potential to address wounds at the molecular and cellular levels. These regenerative approaches target common healing obstacles, such as hyper-inflammation thereby promoting self-recovery through crucial signaling pathway stimulation. The rationale of this review is to examine the benefits and limitations of both current and emerging technologies in wound care and to offer insights into potential advancements in the field. The shift towards such patient-centric therapies reflects a paradigmatic change in wound care strategies.
Collapse
Affiliation(s)
- Yashvi Sharma
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Subhadip Ghatak
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- McGowan Institute of Regenerative Medicine, Department of Surgery, University of Pittsburgh, 419 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Chandan K Sen
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- McGowan Institute of Regenerative Medicine, Department of Surgery, University of Pittsburgh, 419 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| | - Sujata Mohanty
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India.
| |
Collapse
|
29
|
Wang X, Chen S, Chen X, Wu J, Huang Z, Wang J, Chen F, Liu C. Biomimetic multi-channel nerve conduits with micro/nanostructures for rapid nerve repair. Bioact Mater 2024; 41:577-596. [PMID: 39257673 PMCID: PMC11384339 DOI: 10.1016/j.bioactmat.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/13/2024] [Accepted: 07/13/2024] [Indexed: 09/12/2024] Open
Abstract
Peripheral nervous system (PNS) injuries often lead to significant sensory and motor impairments. Traditional artificial nerve conduits, lacking anisotropic structures, have been associated with prolonged repair time and failures in nerve regeneration. This study aimed to address these challenges by developing a novel approach for rapid repair of peripheral nerve injuries (PNI). A 3D oriented fibers scaffold featuring distinct radial (RFs) and longitudinal (LFs) fibers orientations was engineered using coaxial electrospinning and gas directional foaming techniques. This scaffold was then integrated with a shape memory conduit to form a directional multi-channel nerve conduit with micro/nanostructures. The results revealed that the grooved surface of the fibers significantly improved cellular directional guidance, effectively facilitating the migration of SCs from the periphery towards the center and from the base to the apex of the scaffold. In a rat model with a 10 mm nerve defect, the ND-PLATMC/LF ND-PCL scaffold significantly enhanced nerve regeneration and motor function recovery within 4 weeks. These results suggest the potential of this innovative scaffold for efficient repair of the nerve injuries.
Collapse
Affiliation(s)
- Xinqing Wang
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Shuo Chen
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xiaolei Chen
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Juan Wu
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Zhenhua Huang
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jing Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Fangping Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
30
|
Heinzelmann E, Piraino F, Costa M, Roch A, Norkin M, Garnier V, Homicsko K, Brandenberg N. iPSC-derived and Patient-Derived Organoids: Applications and challenges in scalability and reproducibility as pre-clinical models. Curr Res Toxicol 2024; 7:100197. [PMID: 40276485 PMCID: PMC12020925 DOI: 10.1016/j.crtox.2024.100197] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 04/26/2025] Open
Abstract
Recent advancements in stem cell technology have led to the development of organoids - three-dimensional (3D) cell cultures that closely mimic the structural and functional characteristics of human organs. These organoids represent a significant improvement over traditional two-dimensional (2D) cell cultures by preserving native tissue architecture and cellular interactions critical for physiological relevance. This review provides a comprehensive comparison between two main types of organoids: induced Pluripotent Stem Cell (iPSC)-derived and Adult Stem Cell (ASC)-derived (also known as Patient-Derived Organoids, PDOs). iPSC-derived organoids, derived from reprogrammed cells, exhibit remarkable plasticity, and can model a wide range of tissues and developmental stages. They are particularly valuable for studying early human development, genetic disorders, and complex diseases. However, challenges such as prolonged differentiation protocols and variability in maturation levels remain significant hurdles. In contrast, ASC-derived organoids, generated directly from patient tissues, faithfully recapitulate tissue-specific characteristics and disease phenotypes. This fidelity makes them indispensable for personalized medicine applications, including drug screening, disease modeling, and understanding individualized treatment responses. The review highlights the unique advantages and limitations of each organoid type, emphasizing their roles in advancing biomedical research and drug discovery. It addresses key challenges in organoid technology, such as scalability, reproducibility, and the need for standardized culture protocols. Furthermore, it explores recent innovations in scaffold-guided organoid engineering and the integration of organoids with advanced technologies like artificial intelligence and high-throughput screening. The integration of organoids with cutting-edge technologies holds promise for enhancing their utility in modeling complex human diseases and accelerating drug discovery and development. By providing more physiologically relevant models of human organs, organoid technology is poised to revolutionize biomedical research, offering new insights into disease mechanisms and personalized therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Maxim Norkin
- Department of Oncology, CHUV, Lausanne, Switzerland
| | | | | | | |
Collapse
|
31
|
Sun W, Taylor CS, Gao Z, Gregory DA, Haycock JW, Zhao X. Co-assembling bioactive short peptide nanofibers coated silk scaffolds induce neurite outgrowth of PC12 cells. Int J Biol Macromol 2024; 278:134774. [PMID: 39154681 DOI: 10.1016/j.ijbiomac.2024.134774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
Controlling biomolecular-cell interactions is crucial for the design of scaffolds for tissue engineering (TE). Regenerated silk fibroin (RSF) has been extensively used as TE scaffolds, however, RSF showed poor attachment of neuronal cells, such as rat pheochromocytoma (PC12) cells. In this work, amphiphilic peptides containing a hydrophobic isoleucine tail (I3) and laminin or fibronectin derived peptides (IKVAV, PDSGR, YIGSR, RGDS and PHSRN) were designed for promoting scaffold-cell interaction. Three of them (I3KVAV, I3RGDS and I3YIGSR) can self-assemble into nanofibers, therefore, were used to enhance the application of RSF in neuron TE. Live / dead assays revealed that the peptides exhibited negligible cytotoxicity against PC12 cells. The specific interaction between PC12 cells and the peptides were investigate using atomic force microscopy (AFM). The results indicated a synergistic effect in the designed peptides, promoting cellular attachment, proliferation and morphology changes. In addition, AFM results showed that co-assembling peptides I3KVAV and I3YIGSR possesses the best regulation of proliferation and attachment of PC12 cells, consistent with immunofluorescence staining results. Moreover, cell culture with hydrogels revealed that a mixture of peptides I3KVAV and I3YIGSR can also promote 3D neurites outgrowth. The approach of combining two different self-assembling peptides shows great potential for nerve regeneration applications.
Collapse
Affiliation(s)
- Weizhen Sun
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - Caroline S Taylor
- Department of Materials Science & Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - Zijian Gao
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - David A Gregory
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - John W Haycock
- Department of Materials Science & Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK.
| |
Collapse
|
32
|
Wang Y, Liu M, Zhang W, Liu H, Jin F, Mao S, Han C, Wang X. Mechanical strategies to promote vascularization for tissue engineering and regenerative medicine. BURNS & TRAUMA 2024; 12:tkae039. [PMID: 39350780 PMCID: PMC11441985 DOI: 10.1093/burnst/tkae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 10/04/2024]
Abstract
Vascularization is a major challenge in the field of tissue engineering and regenerative medicine. Mechanical factors have been demonstrated to play a fundamental role in vasculogenesis and angiogenesis and can affect the architecture of the generated vascular network. Through the regulation of mechanical factors in engineered tissues, various mechanical strategies can be used to optimize the preformed vascular network and promote its rapid integration with host vessels. Optimization of the mechanical properties of scaffolds, including controlling scaffold stiffness, increasing surface roughness and anisotropic structure, and designing interconnected, hierarchical pore structures, is beneficial for the in vitro formation of vascular networks and the ingrowth of host blood vessels. The incorporation of hollow channels into scaffolds promotes the formation of patterned vascular networks. Dynamic stretching and perfusion can facilitate the formation and maturation of preformed vascular networks in vitro. Several indirect mechanical strategies provide sustained mechanical stimulation to engineered tissues in vivo, which further promotes the vascularization of implants within the body. Additionally, stiffness gradients, anisotropic substrates and hollow channels in scaffolds, as well as external cyclic stretch, boundary constraints and dynamic flow culture, can effectively regulate the alignment of vascular networks, thereby promoting better integration of prevascularized engineered tissues with host blood vessels. This review summarizes the influence and contribution of both scaffold-based and external stimulus-based mechanical strategies for vascularization in tissue engineering and elucidates the underlying mechanisms involved.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Meixuan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Wei Zhang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Huan Liu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Fang Jin
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Shulei Mao
- Department of Burns and Plastic Surgery, Quhua Hospital of Zhejiang, 62 Wenchang Road, Quhua, Quzhou 324004, China
| | - Chunmao Han
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| | - Xingang Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, 88 Jiefang Road, Shangcheng District, Hangzhou 310009, China
| |
Collapse
|
33
|
Hou Y, Conklin B, Choi HK, Yang L, Lee KB. Probing Nanotopography-Mediated Macrophage Polarization via Integrated Machine Learning and Combinatorial Biophysical Cue Mapping. ACS NANO 2024; 18:25465-25477. [PMID: 39226301 DOI: 10.1021/acsnano.4c04406] [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] [Indexed: 09/05/2024]
Abstract
Inflammatory responses, leading to fibrosis and potential host rejection, significantly hinder the long-term success and widespread adoption of biomedical implants. The ability to control and investigated macrophage inflammatory responses at the implant-macrophage interface would be critical for reducing chronic inflammation and improving tissue integration. Nonetheless, the systematic investigation of how surface topography affects macrophage polarization is typically complicated by the restricted complexity of accessible nanostructures, difficulties in achieving exact control, and biased preselection of experimental parameters. In response to these problems, we developed a large-scale, high-content combinatorial biophysical cue (CBC) array for enabling high-throughput screening (HTS) of the effects of nanotopography on macrophage polarization and subsequent inflammatory processes. Our CBC array, created utilizing the dynamic laser interference lithography (DLIL) technology, contains over 1 million nanotopographies, ranging from nanolines and nanogrids to intricate hierarchical structures with dimensions ranging from 100 nm to several microns. Using machine learning (ML) based on the Gaussian process regression algorithm, we successfully identified certain topographical signals that either repress (pro-M2) or stimulate (pro-M1) macrophage polarization. The upscaling of these nanotopographies for further examination has shown mechanisms such as cytoskeletal remodeling and ROCK-dependent epigenetic activation to be critical to the mechanotransduction pathways regulating macrophage fate. Thus, we have also developed a platform combining advanced DLIL nanofabrication techniques, HTS, ML-driven prediction of nanobio interactions, and mechanotransduction pathway evaluation. In short, our developed platform technology not only improves our ability to investigate and understand nanotopography-regulated macrophage inflammatory responses but also holds great potential for guiding the design of nanostructured coatings for therapeutic biomaterials and biomedical implants.
Collapse
Affiliation(s)
- Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Brandon Conklin
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Hye Kyu Choi
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Science and Technology, Tongji University, Shanghai 200065, China
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
34
|
Su Z, Xing Y, Xiao Y, Guo J, Wang C, Wang F, Xu Z, Wu W, Gu Y. Decellularized, Heparinized Small-Caliber Tissue-Engineered "Biological Tubes" for Allograft Vascular Grafts. ACS Biomater Sci Eng 2024; 10:5154-5167. [PMID: 39079153 DOI: 10.1021/acsbiomaterials.4c00634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
There remains a lack of small-caliber tissue-engineered blood vessels (TEBVs) with wide clinical use. Biotubes were developed by electrospinning and in-body tissue architecture (iBTA) technology to prepare small-caliber TEBVs with promising applications. Different ratios of hybrid fibers of poly(l-lactic-co-ε-caprolactone) (PLCL) and polyurethane (PU) were obtained by electrospinning, and the electrospun tubes were then implanted subcutaneously in the abdominal area of a rabbit (as an in vivo bioreactor). The biotubes were harvested after 4 weeks. They were then decellularized and cross-linked with heparin. PLCL/PU electrospun vascular tubes, decellularized biotubes (D-biotubes), and heparinized combined decellularized biotubes (H + D-biotubes) underwent carotid artery allograft transplantation in a rabbit model. Vascular ultrasound follow-up and histological observation revealed that the biotubes developed based on electrospinning and iBTA technology, after decellularization and heparinization cross-linking, showed a better patency rate, adequate mechanical properties, and remodeling ability in the rabbit model. IBTA technology caused a higher patency, and the heparinization cross-linking process gave the biotubes stronger mechanical properties.
Collapse
Affiliation(s)
- Zhixiang Su
- Vascular Surgery Department, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218 Beijing, China
| | - Yuehao Xing
- Department of Cardiovascular Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045 Beijing, China
| | - Yonghao Xiao
- School of Materials Science and Engineering, Beijing Institute of Technology, 100086 Beijing, China
| | - Julong Guo
- Vascular Surgery Department, Xuanwu Hospital, Capital Medical University, 100053 Beijing, China
| | - Cong Wang
- Vascular Surgery Department, Xuanwu Hospital, Capital Medical University, 100053 Beijing, China
| | - Fei Wang
- Vascular Surgery Department, Xuanwu Hospital, Capital Medical University, 100053 Beijing, China
| | - Zeqin Xu
- Vascular Surgery Department, Xuanwu Hospital, Capital Medical University, 100053 Beijing, China
| | - Weiwei Wu
- Vascular Surgery Department, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218 Beijing, China
| | - Yongquan Gu
- Vascular Surgery Department, Xuanwu Hospital, Capital Medical University, 100053 Beijing, China
| |
Collapse
|
35
|
Yang Y, Zhang X, Yan H, Zhao R, Zhang R, Zhu L, Zhang J, Midgley AC, Wan Y, Wang S, Qian M, Zhao Q, Ai D, Wang T, Kong D, Huang X, Wang K. Versatile Design of NO-Generating Proteolipid Nanovesicles for Alleviating Vascular Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401844. [PMID: 38884204 PMCID: PMC11336937 DOI: 10.1002/advs.202401844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/23/2024] [Indexed: 06/18/2024]
Abstract
Vascular injury is central to the pathogenesis and progression of cardiovascular diseases, however, fostering alternative strategies to alleviate vascular injury remains a persisting challenge. Given the central role of cell-derived nitric oxide (NO) in modulating the endogenous repair of vascular injury, NO-generating proteolipid nanovesicles (PLV-NO) are designed that recapitulate the cell-mimicking functions for vascular repair and replacement. Specifically, the proteolipid nanovesicles (PLV) are versatilely fabricated using membrane proteins derived from different types of cells, followed by the incorporation of NO-generating nanozymes capable of catalyzing endogenous donors to produce NO. Taking two vascular injury models, two types of PLV-NO are tailored to meet the individual requirements of targeted diseases using platelet membrane proteins and endothelial membrane proteins, respectively. The platelet-based PLV-NO (pPLV-NO) demonstrates its efficacy in targeted repair of a vascular endothelium injury model through systemic delivery. On the other hand, the endothelial cell (EC)-based PLV-NO (ePLV-NO) exhibits suppression of thrombosis when modified onto a locally transplanted small-diameter vascular graft (SDVG). The versatile design of PLV-NO may enable a promising therapeutic option for various vascular injury-evoked cardiovascular diseases.
Collapse
Affiliation(s)
- Yueyue Yang
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Xiangyun Zhang
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Hongyu Yan
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Rongping Zhao
- School of MedicineNankai UniversityTianjin300071China
| | - Ruixin Zhang
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Liuyang Zhu
- First Central Clinical CollegeTianjin Medical UniversityTianjin300192China
| | - Jingai Zhang
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Adam C. Midgley
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Ye Wan
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Songdi Wang
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Meng Qian
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Qiang Zhao
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Ding Ai
- Department of Physiology and PathophysiologyTianjin Medical UniversityTianjin300070China
| | - Ting Wang
- Tianjin Key Laboratory of Urban Transport Emission ResearchCollege of Environmental Science and EngineeringNankai UniversityTianjin300071China
| | - Deling Kong
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Kai Wang
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| |
Collapse
|
36
|
Garg R, Gonuguntla S, Sk S, Iqbal MS, Dada AO, Pal U, Ahmadipour M. Sputtering thin films: Materials, applications, challenges and future directions. Adv Colloid Interface Sci 2024; 330:103203. [PMID: 38820883 DOI: 10.1016/j.cis.2024.103203] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 06/02/2024]
Abstract
Sputtering is an effective technique for producing ultrathin films with diverse applications. The review begins by providing an in-depth overview of the background, introducing the early development of sputtering and its principles. Consequently, progress in advancements made in recent decades highlights the renaissance of sputtering as a powerful technology for creating thin films with varied compositions, structures, and properties. For the first time, we have discussed a thorough overview of several sputtered thin film materials based on metal and metal oxide, metal nitride, alloys, carbon, and ceramic-based thin film along with their properties and their applicability in various fields. We further delve into the applications of sputter-coated thin films, specifically emphasizing their relevance in environmental sustainability, energy and electronics, and biomedical fields. We critically examine the recent advancements in developing sputter-coated catalysts for eliminating water pollutants andhydrogen generation. Additionally, the review sheds light on advantages, shortcomings, and future directions for developing sputter-coated thin films utilized in biodegradable metals and alloys with enhanced corrosion resistance and biocompatibility. This review is a comprehensive integration of recent literature, covering diverse sputtering thin film applications. We delve deeply into various material types and emphasize critical analysis of recent advancements, particularly in environmental, energy, and biomedical fields. By offering insights into both advancements and limitations, the review provides a nuanced understanding essential for practical utilization.
Collapse
Affiliation(s)
- Renuka Garg
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, PO Box 26666, United Arab Emirates
| | - Spandana Gonuguntla
- Department of Energy & Environmental Engineering, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Saddam Sk
- Department of Energy & Environmental Engineering, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Muhammad Saqlain Iqbal
- Department of Chemistry, COMSATS University Islamabad, Lahore campus, 54000 Lahore, Pakistan
| | - Adewumi Oluwasogo Dada
- Department of Energy & Environmental Engineering, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Industrial Chemistry Programme, Nanotechnology Laboratory, Department of Physical Sciences, Landmark University, P.M.B.1001, Omu-Aran, Kwara, Nigeria
| | - Ujjwal Pal
- Department of Energy & Environmental Engineering, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Mohsen Ahmadipour
- Institute of Power Engineering, Universiti Tenaga Nasional, Serdang, Malaysia.
| |
Collapse
|
37
|
Kong L, Gao X, Yao X, Xie H, Kang Q, Sun W, You Z, Qian Y, Fan C. Multilevel neurium-mimetic individualized graft via additive manufacturing for efficient tissue repair. Nat Commun 2024; 15:6428. [PMID: 39079956 PMCID: PMC11289102 DOI: 10.1038/s41467-024-49980-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Complicated peripheral nerve injuries or defects, especially at branching sites, remain a prominent clinical challenge after the application of different treatment strategies. Current nerve grafts fail to match the expected shape and size for delicate and precise branched nerve repair on a case-by-case basis, and there is a lack of geometrical and microscale regenerative navigation. In this study, we develop a sugar painting-inspired individualized multilevel epi-/peri-/endoneurium-mimetic device (SpinMed) to customize natural cues, featuring a selectively protective outer sheath and an instructive core, to support rapid vascular reconstruction and consequent efficient neurite extension along the defect area. The biomimetic perineurium dictates host-guest crosslinking in which new vessels secrete multimerin 1 binding to the fibroin filler surface as an anchor, contributing to the biological endoneurium that promotes Schwann cell homing and remyelination. SpinMed implantation into rat sciatic nerve defects yields a satisfactory outcome in terms of structural reconstruction, with sensory and locomotive function restoration. We further customize SpinMed grafts based on anatomy and digital imaging, achieving rapid repair of the nerve trunk and branches superior to that achieved by autografts and decellularized grafts in a specific beagle nerve defect model, with reliable biosafety. Overall, this intelligent art-inspired biomimetic design offers a facile way to customize sophisticated high-performance nerve grafts and holds great potential for application in translational regenerative medicine.
Collapse
Affiliation(s)
- Lingchi Kong
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Xin Gao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, 201620, Shanghai, China
| | - Xiangyun Yao
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, 201306, Shanghai, China
| | - Haijiao Xie
- Hangzhou Yanqu Information Technology Co.Ltd., 310003, Hangzhou, China
| | - Qinglin Kang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Wei Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, 201620, Shanghai, China.
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, 201620, Shanghai, China.
| | - Yun Qian
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China.
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, 201306, Shanghai, China.
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China.
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, 201306, Shanghai, China.
| |
Collapse
|
38
|
Alizadeh S, Ameri Z, Daemi H, Pezeshki-Modaress M. Sulfated polysaccharide as biomimetic biopolymers for tissue engineering scaffolds fabrication: Challenges and opportunities. Carbohydr Polym 2024; 336:122124. [PMID: 38670755 DOI: 10.1016/j.carbpol.2024.122124] [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: 12/14/2023] [Revised: 03/31/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024]
Abstract
Sulfated polysaccharides play important roles in tissue engineering applications because of their high growth factor preservation ability and their native-like biological features. There are different sulfated polysaccharides based on different repeating units in the carbohydrate backbone, the position of the sulfate group, and the sulfation degree of the polysaccharide. These led to various sulfated polymers with different negative charge densities and resultant structure-property relationships. Since numerous reports are presented related to sulfated polysaccharide applications in tissue engineering, it is crucial to review the role of effective physicochemical and biological parameters in their usage; as well as their structure-property relationships. Within this review, we focused on the effect of naturally occurring and synthetic sulfated polysaccharides in tissue engineering applications reported in the last years, highlighting the challenges of the scaffold fabrication process, the position, and the degree of sulfate on biomedical activity. Additionally, we discussed their use in numerous in vitro and in vivo model systems.
Collapse
Affiliation(s)
- Sanaz Alizadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Ameri
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamed Daemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamad Pezeshki-Modaress
- Burn Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Plastic and Reconstructive surgery, Hazrat Fatemeh Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Huang T, Zeng Y, Li C, Zhou Z, Xu J, Wang L, Yu DG, Wang K. Application and Development of Electrospun Nanofiber Scaffolds for Bone Tissue Engineering. ACS Biomater Sci Eng 2024; 10:4114-4144. [PMID: 38830819 DOI: 10.1021/acsbiomaterials.4c00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Nanofiber scaffolds have gained significant attention in the field of bone tissue engineering. Electrospinning, a straightforward and efficient technique for producing nanofibers, has been extensively researched. When used in bone tissue engineering scaffolds, electrospun nanofibers with suitable surface properties promote new bone tissue growth and enhance cell adhesion. Recent advancements in electrospinning technology have provided innovative approaches for scaffold fabrication in bone tissue engineering. This review comprehensively examines the utilization of electrospun nanofibers in bone tissue engineering scaffolds and evaluates the relevant literature. The review begins by presenting the fundamental principles and methodologies of electrospinning. It then discusses various materials used in the production of electrospun nanofiber scaffolds for bone tissue engineering, including natural and synthetic polymers, as well as certain inorganic materials. The challenges associated with these materials are also described. The review focuses on novel electrospinning techniques for scaffold construction in bone tissue engineering, such as multilayer nanofibers, multifluid electrospinning, and the integration of electrospinning with other methods. Recent advancements in electrospinning technology have enabled the fabrication of precisely aligned nanofiber scaffolds with nanoscale architectures. These innovative methods also facilitate the fabrication of biomimetic structures, wherein bioactive substances can be incorporated and released in a controlled manner for drug delivery purposes. Moreover, they address issues encountered with traditional electrospun nanofibers, such as mechanical characteristics and biocompatibility. Consequently, the development and implementation of novel electrospinning technologies have revolutionized scaffold fabrication for bone tissue engineering.
Collapse
Affiliation(s)
- Tianyue Huang
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| | - YuE Zeng
- Department of Neurology, RuiJin Hospital Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chaofei Li
- Department of General Surgery, RuiJin Hospital Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhengqing Zhou
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| | - Jie Xu
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| | - Lean Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| | - Deng-Guang Yu
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| | - Ke Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| |
Collapse
|
40
|
陈 明, 吴 江, 殷 瀚, 眭 翔, 刘 舒, 郭 全. [Construction of a novel tissue engineered meniscus scaffold based on low temperature deposition three-dimenisonal printing technology]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2024; 38:748-754. [PMID: 38918198 PMCID: PMC11190681 DOI: 10.7507/1002-1892.202402063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024]
Abstract
Objective To investigate the construction of a novel tissue engineered meniscus scaffold based on low temperature deposition three-dimenisonal (3D) printing technology and evaluate its biocompatibility. Methods The fresh pig meniscus was decellularized by improved physicochemical method to obtain decellularized meniscus matrix homogenate. Gross observation, HE staining, and DAPI staining were used to observe the decellularization effect. Toluidine blue staining, safranin O staining, and sirius red staining were used to evaluate the retention of mucopolysaccharide and collagen. Then, the decellularized meniscus matrix bioink was prepared, and the new tissue engineered meniscus scaffold was prepared by low temperature deposition 3D printing technology. Scanning electron microscopy was used to observe the microstructure. After co-culture with adipose-derived stem cells, the cell compatibility of the scaffolds was observed by cell counting kit 8 (CCK-8), and the cell activity and morphology were observed by dead/live cell staining and cytoskeleton staining. The inflammatory cell infiltration and degradation of the scaffolds were evaluated by subcutaneous experiment in rats. Results The decellularized meniscus matrix homogenate appeared as a transparent gel. DAPI and histological staining showed that the immunogenic nucleic acids were effectively removed and the active components of mucopolysaccharide and collagen were remained. The new tissue engineered meniscus scaffolds was constructed by low temperature deposition 3D printing technology and it had macroporous-microporous microstructures under scanning electron microscopy. CCK-8 test showed that the scaffolds had good cell compatibility. Dead/live cell staining showed that the scaffold could effectively maintain cell viability (>90%). Cytoskeleton staining showed that the scaffolds were benefit for cell adhesion and spreading. After 1 week of subcutaneous implantation of the scaffolds in rats, there was a mild inflammatory response, but no significant inflammatory response was observed after 3 weeks, and the scaffolds gradually degraded. Conclusion The novel tissue engineered meniscus scaffold constructed by low temperature deposition 3D printing technology has a graded macroporous-microporous microstructure and good cytocompatibility, which is conducive to cell adhesion and growth, laying the foundation for the in vivo research of tissue engineered meniscus scaffolds in the next step.
Collapse
Affiliation(s)
- 明学 陈
- 首都医科大学附属北京积水潭医院矫形骨科(北京 100035)Department of Orthopaedics, Beijing Jishuitan Hospital Affiliated to Capital Medical University, Beijing, 100035, P. R. China
| | - 江 吴
- 首都医科大学附属北京积水潭医院矫形骨科(北京 100035)Department of Orthopaedics, Beijing Jishuitan Hospital Affiliated to Capital Medical University, Beijing, 100035, P. R. China
- 中国人民解放军总医院第一医学中心骨科研究所 骨科再生医学北京市重点实验室 全军骨科战创伤重点实验室(北京 100853)Institute of Orthopedics, the First Medical Center of the Chinese PLA General Hospital, Beijing Key Laboratory of Orthopaedic Regenerative Medicine, Key Laboratory of Orthopaedic War Trauma, Beijing, 100853, P. R. China
| | - 瀚 殷
- 首都医科大学附属北京积水潭医院矫形骨科(北京 100035)Department of Orthopaedics, Beijing Jishuitan Hospital Affiliated to Capital Medical University, Beijing, 100035, P. R. China
| | - 翔 眭
- 首都医科大学附属北京积水潭医院矫形骨科(北京 100035)Department of Orthopaedics, Beijing Jishuitan Hospital Affiliated to Capital Medical University, Beijing, 100035, P. R. China
| | - 舒云 刘
- 首都医科大学附属北京积水潭医院矫形骨科(北京 100035)Department of Orthopaedics, Beijing Jishuitan Hospital Affiliated to Capital Medical University, Beijing, 100035, P. R. China
| | - 全义 郭
- 首都医科大学附属北京积水潭医院矫形骨科(北京 100035)Department of Orthopaedics, Beijing Jishuitan Hospital Affiliated to Capital Medical University, Beijing, 100035, P. R. China
| |
Collapse
|
41
|
Wang X, Li K, Yuan Y, Zhang N, Zou Z, Wang Y, Yan S, Li X, Zhao P, Li Q. Nonlinear Elasticity of Blood Vessels and Vascular Grafts. ACS Biomater Sci Eng 2024; 10:3631-3654. [PMID: 38815169 DOI: 10.1021/acsbiomaterials.4c00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
The transplantation of vascular grafts has emerged as a prevailing approach to address vascular disorders. However, the development of small-diameter vascular grafts is still in progress, as they serve in a more complicated mechanical environment than their counterparts with larger diameters. The biocompatibility and functional characteristics of small-diameter vascular grafts have been well developed; however, mismatch in mechanical properties between the vascular grafts and native arteries has not been accomplished, which might facilitate the long-term patency of small-diameter vascular grafts. From a point of view in mechanics, mimicking the nonlinear elastic mechanical behavior exhibited by natural blood vessels might be the state-of-the-art in designing vascular grafts. This review centers on elucidating the nonlinear elastic behavior of natural blood vessels and vascular grafts. The biological functionality and limitations associated with as-reported vascular grafts are meticulously reviewed and the future trajectory for fabricating biomimetic small-diameter grafts is discussed. This review might provide a different insight from the traditional design and fabrication of artificial vascular grafts.
Collapse
Affiliation(s)
- Xiaofeng Wang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
- The State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Kecheng Li
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yuan Yuan
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Ning Zhang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Zifan Zou
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yun Wang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Shujie Yan
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Peng Zhao
- The State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Qian Li
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
42
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
43
|
Wang J, Yang X, Xu M, Liu H, Liu L, Tan Z. Distinct cellular microenvironment with cytotypic effects regulates orderly regeneration of vascular tissues. Mater Today Bio 2024; 26:101033. [PMID: 38533377 PMCID: PMC10963652 DOI: 10.1016/j.mtbio.2024.101033] [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: 12/05/2023] [Revised: 02/26/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
Regeneration of the architecturally complex blood vascular system requires precise temporal and spatial control of cell behaviours. Additional components must be integrated into the structure to achieve clinical success for in situ tissue engineering. Consequently, this study proposed a universal method for including any substrate type in vascular cell extracellular matrices (VCEM) via regulating selective adhesion to promote vascular tissue regeneration. The results uncovered that the VCEM worked as cell adhesion substrates, exhibited cell type specificity, and functioned as an address signal for recognition by vascular cells, which resulted in matching with the determined cells. The qPCR and immunofluorescence results revealed that a cell type-specific VCEM could be designed to promote or inhibit cell adhesion, consistenting with the expression patterns of eyes absent 3 (Eya3). In addition, a 3D vascular graft combined with VCEM which could recapitulate the vascular cell-like microenvironment was fabricated. The vascular graft revealed a prospective role for cellular microenvironment in the establishment of vascular cell distribution and tissue architecture, and potentiated the orderly regeneration and functional recovery of vascular tissues in vivo. The findings demonstrate that differential adhesion between cell types due to the cellular microenvironment is sufficient to drive the complex assembly of engineered blood vessel functional units, and underlies hierarchical organization during vascular regeneration.
Collapse
Affiliation(s)
- Jian Wang
- College of Biology, Hunan University, Changsha, 410082, China
- Institute of Shenzhen, Hunan University Shenzhen, 518000, China
| | - Xun Yang
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital, Shenzhen University), Shenzhen, 518028, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Miaomiao Xu
- College of Biology, Hunan University, Changsha, 410082, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| | - Hui Liu
- College of Biology, Hunan University, Changsha, 410082, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| | - Lijun Liu
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital, Shenzhen University), Shenzhen, 518028, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha, 410082, China
- Institute of Shenzhen, Hunan University Shenzhen, 518000, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| |
Collapse
|
44
|
Hu N, Jiang R, Deng Y, Li W, Jiang W, Xu N, Wang J, Wen J, Gu S. Periapical lesion-derived decellularized extracellular matrix as a potential solution for regenerative endodontics. Regen Biomater 2024; 11:rbae050. [PMID: 38872841 PMCID: PMC11170217 DOI: 10.1093/rb/rbae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 04/01/2024] [Accepted: 04/17/2024] [Indexed: 06/15/2024] Open
Abstract
Pulp regeneration remains a crucial target in the preservation of natural dentition. Using decellularized extracellular matrix is an appropriate approach to mimic natural microenvironment and facilitate tissue regeneration. In this study, we attempted to obtain decellularized extracellular matrix from periapical lesion (PL-dECM) and evaluate its bioactive effects. The decellularization process yielded translucent and viscous PL-dECM, meeting the standard requirements for decellularization efficiency. Proteomic sequencing revealed that the PL-dECM retained essential extracellular matrix components and numerous bioactive factors. The PL-dECM conditioned medium could enhance the proliferation and migration ability of periapical lesion-derived stem cells (PLDSCs) in a dose-dependent manner. Culturing PLDSCs on PL-dECM slices improved odontogenic/angiogenic ability compared to the type I collagen group. In vivo, the PL-dECM demonstrated a sustained supportive effect on PLDSCs and promoted odontogenic/angiogenic differentiation. Both in vitro and in vivo studies illustrated that PL-dECM served as an effective scaffold for pulp tissue engineering, providing valuable insights into PLDSCs differentiation. These findings pave avenues for the clinical application of dECM's in situ transplantation for regenerative endodontics.
Collapse
Affiliation(s)
- Nan Hu
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No.639, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Yanqiao Road No.390, Shanghai, 200125, China
- National Center for Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Zhizaoju Road No.639, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Yanqiao Road No.390, Shanghai, 200125, China
- Shanghai Research Institute of Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
| | - Ruixue Jiang
- Department of Prosthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No.639, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Yanqiao Road No.390, Shanghai, 200125, China
- National Center for Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Zhizaoju Road No.639, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Yanqiao Road No.390, Shanghai, 200125, China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Yanqiao Road No.390, Shanghai, 200125, China
| | - Yuwei Deng
- Department of Prosthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No.639, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Yanqiao Road No.390, Shanghai, 200125, China
- National Center for Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Zhizaoju Road No.639, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Yanqiao Road No.390, Shanghai, 200125, China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Yanqiao Road No.390, Shanghai, 200125, China
| | - Weiping Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No.639, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Yanqiao Road No.390, Shanghai, 200125, China
- National Center for Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Zhizaoju Road No.639, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Yanqiao Road No.390, Shanghai, 200125, China
- Shanghai Research Institute of Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Zhizaoju Road No.639, Shanghai, 200011, China
| | - Wentao Jiang
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No.639, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Yanqiao Road No.390, Shanghai, 200125, China
- National Center for Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Zhizaoju Road No.639, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Yanqiao Road No.390, Shanghai, 200125, China
- Shanghai Research Institute of Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
| | - Ningwei Xu
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No.639, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Yanqiao Road No.390, Shanghai, 200125, China
- National Center for Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Zhizaoju Road No.639, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Yanqiao Road No.390, Shanghai, 200125, China
- Shanghai Research Institute of Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
| | - Jia Wang
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No.639, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Yanqiao Road No.390, Shanghai, 200125, China
- National Center for Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Zhizaoju Road No.639, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Yanqiao Road No.390, Shanghai, 200125, China
- Shanghai Research Institute of Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
| | - Jin Wen
- Department of Prosthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No.639, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Yanqiao Road No.390, Shanghai, 200125, China
- National Center for Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Zhizaoju Road No.639, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Yanqiao Road No.390, Shanghai, 200125, China
- Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Yanqiao Road No.390, Shanghai, 200125, China
| | - Shensheng Gu
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No.639, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Yanqiao Road No.390, Shanghai, 200125, China
- National Center for Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Zhizaoju Road No.639, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Yanqiao Road No.390, Shanghai, 200125, China
- Shanghai Research Institute of Stomatology, Zhizaoju Road No.639, Shanghai, 200011, China
| |
Collapse
|
45
|
Wei S, Li Z, Xia H, Wang Z, Deng J, Li L, Huang R, Ye T, Huang Y, Yang Y. An endometrial biomimetic extracellular matrix (ECM) for enhanced endometrial regeneration using hyaluronic acid hydrogel containing recombinant human type III collagen. Int J Biol Macromol 2024; 268:131723. [PMID: 38649072 DOI: 10.1016/j.ijbiomac.2024.131723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Endometrial injury poses a significant challenge in tissue regeneration, with type III collagen (COL III) playing a pivotal role in maintaining endometrial integrity and facilitating repair. Our study explored the utility of recombinant human type III collagen (RHC) as an intervention for endometrial damage. To address the challenges associated with the inherent instability and rapid degradation of COL III in vivo, we developed an RHC-HA hydrogel by conjugating RHC with hyaluronic acid (HA), thus ensuring a more stable and sustained delivery. Our findings suggested that the RHC-HA hydrogel significantly promoted endometrial regeneration and restored fertility. The hydrogel facilitated prolonged retention of RHC in the uterus, leading to a substantial improvement in the repair process. The synergistic interaction between RHC and HA greatly enhances cell proliferation and adhesion, surpassing the efficacy of HA or RHC alone. Additionally, the RHC-HA hydrogel demonstrated notable anti-fibrotic effects, which are crucial for preventing abnormalities during endometrial healing. These findings suggested that the RHC-HA hydrogel presented a therapeutic strategy in the treatment of uterine endometrial injuries, which may improve female reproductive health.
Collapse
Affiliation(s)
- Siying Wei
- Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Ziyi Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Huan Xia
- Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Zhaoyang Wang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Jingxian Deng
- Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Lu Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Rufei Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Tao Ye
- Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Yadong Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China.
| | - Yan Yang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China.
| |
Collapse
|
46
|
Rahmati S, Khazaei M, Abpeikar Z, Soleimanizadeh A, Rezakhani L. Exosome-loaded decellularized tissue: Opening a new window for regenerative medicine. J Tissue Viability 2024; 33:332-344. [PMID: 38594147 DOI: 10.1016/j.jtv.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 03/29/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
Mesenchymal stem cell-derived exosomes (MSCs-EXO) have received a lot of interest recently as a potential therapeutic tool in regenerative medicine. Extracellular vesicles (EVs) known as exosomes (EXOs) are crucial for cell-cell communication throughout a variety of activities including stress response, aging, angiogenesis, and cell differentiation. Exploration of the potential use of EXOs as essential therapeutic effectors of MSCs to encourage tissue regeneration was motivated by success in the field of regenerative medicine. EXOs have been administered to target tissues using a variety of methods, including direct, intravenous, intraperitoneal injection, oral delivery, and hydrogel-based encapsulation, in various disease models. Despite the significant advances in EXO therapy, various methods are still being researched to optimize the therapeutic applications of these nanoparticles, and it is not completely clear which approach to EXO administration will have the greatest effects. Here, we will review emerging developments in the applications of EXOs loaded into decellularized tissues as therapeutic agents for use in regenerative medicine in various tissues.
Collapse
Affiliation(s)
- Shima Rahmati
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Abpeikar
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Arghavan Soleimanizadeh
- Faculty of Medicine, Graduate School 'Molecular Medicine, University of Ulm, 89081, Ulm, Germany
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
47
|
Sharifi M, Kamalabadi-Farahani M, Salehi M, Ebrahimi-Barough S, Alizadeh M. Recent advances in enhances peripheral nerve orientation: the synergy of micro or nano patterns with therapeutic tactics. J Nanobiotechnology 2024; 22:194. [PMID: 38643117 PMCID: PMC11031871 DOI: 10.1186/s12951-024-02475-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/11/2024] [Indexed: 04/22/2024] Open
Abstract
Several studies suggest that topographical patterns influence nerve cell fate. Efforts have been made to improve nerve cell functionality through this approach, focusing on therapeutic strategies that enhance nerve cell function and support structures. However, inadequate nerve cell orientation can impede long-term efficiency, affecting nerve tissue repair. Therefore, enhancing neurites/axons directional growth and cell orientation is crucial for better therapeutic outcomes, reducing nerve coiling, and ensuring accurate nerve fiber connections. Conflicting results exist regarding the effects of micro- or nano-patterns on nerve cell migration, directional growth, immunogenic response, and angiogenesis, complicating their clinical use. Nevertheless, advances in lithography, electrospinning, casting, and molding techniques to intentionally control the fate and neuronal cells orientation are being explored to rapidly and sustainably improve nerve tissue efficiency. It appears that this can be accomplished by combining micro- and nano-patterns with nanomaterials, biological gradients, and electrical stimulation. Despite promising outcomes, the unclear mechanism of action, the presence of growth cones in various directions, and the restriction of outcomes to morphological and functional nerve cell markers have presented challenges in utilizing this method. This review seeks to clarify how micro- or nano-patterns affect nerve cell morphology and function, highlighting the potential benefits of cell orientation, especially in combined approaches.
Collapse
Affiliation(s)
- Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| | | | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Alizadeh
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
48
|
Liao J, Zhou Y, Zhao X, Hou B, Zhang J, Huang H. Chitin microspheres: From fabrication to applications. Carbohydr Polym 2024; 329:121773. [PMID: 38286547 DOI: 10.1016/j.carbpol.2023.121773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/31/2024]
Abstract
Chitin microspheres (CMs) have attracted increasing attention due to their biocompatibility, uniform size and shape, large surface area, and porous structure. Considerable research efforts have been focused on developing CMs and promoting their applications in various areas. In this context, this review aims to describe the most recent progress in the fabrication and application of CMs. Different routes that can be used to prepare CMs, such as the drip method and the emulsion method, are emphatically introduced. Moreover, the applications of CMs as drug delivery systems, wound dressings, three-dimensional (3D) scaffolds, water purification, and functional supporting materials in the fields of biomedicine, tissue engineering, environmental protection, and energy storage are also highlighted. We hope this review can provide a comprehensive and useful database for further innovation of CMs.
Collapse
Affiliation(s)
- Jing Liao
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Meat Processing Key Laboratory of Sichuan Province, Chengdu University, Chengdu 610106, China.
| | - Yuhang Zhou
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Xingyue Zhao
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Bo Hou
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Jiamin Zhang
- Meat Processing Key Laboratory of Sichuan Province, Chengdu University, Chengdu 610106, China.
| | - Huihua Huang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China.
| |
Collapse
|
49
|
Zhou W, Rahman MSU, Sun C, Li S, Zhang N, Chen H, Han CC, Xu S, Liu Y. Perspectives on the Novel Multifunctional Nerve Guidance Conduits: From Specific Regenerative Procedures to Motor Function Rebuilding. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307805. [PMID: 37750196 DOI: 10.1002/adma.202307805] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/19/2023] [Indexed: 09/27/2023]
Abstract
Peripheral nerve injury potentially destroys the quality of life by inducing functional movement disorders and sensory capacity loss, which results in severe disability and substantial psychological, social, and financial burdens. Autologous nerve grafting has been commonly used as treatment in the clinic; however, its rare donor availability limits its application. A series of artificial nerve guidance conduits (NGCs) with advanced architectures are also proposed to promote injured peripheral nerve regeneration, which is a complicated process from axon sprouting to targeted muscle reinnervation. Therefore, exploring the interactions between sophisticated NGC complexes and versatile cells during each process including axon sprouting, Schwann cell dedifferentiation, nerve myelination, and muscle reinnervation is necessary. This review highlights the contribution of functional NGCs and the influence of microscale biomaterial architecture on biological processes of nerve repair. Progressive NGCs with chemical molecule induction, heterogenous topographical morphology, electroactive, anisotropic assembly microstructure, and self-powered electroactive and magnetic-sensitive NGCs are also collected, and they are expected to be pioneering features in future multifunctional and effective NGCs.
Collapse
Affiliation(s)
- Weixian Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Muhammad Saif Ur Rahman
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education Guangdong province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Chengmei Sun
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education Guangdong province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Nuozi Zhang
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Hao Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Charles C Han
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Materials Science and Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Materials Science and Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
50
|
Fu Y, Zhou Y, Wang K, Li Z, Kong W. Extracellular Matrix Interactome in Modulating Vascular Homeostasis and Remodeling. Circ Res 2024; 134:931-949. [PMID: 38547250 DOI: 10.1161/circresaha.123.324055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
The ECM (extracellular matrix) is a major component of the vascular microenvironment that modulates vascular homeostasis. ECM proteins include collagens, elastin, noncollagen glycoproteins, and proteoglycans/glycosaminoglycans. ECM proteins form complex matrix structures, such as the basal lamina and collagen and elastin fibers, through direct interactions or lysyl oxidase-mediated cross-linking. Moreover, ECM proteins directly interact with cell surface receptors or extracellular secreted molecules, exerting matricellular and matricrine modulation, respectively. In addition, extracellular proteases degrade or cleave matrix proteins, thereby contributing to ECM turnover. These interactions constitute the ECM interactome network, which is essential for maintaining vascular homeostasis and preventing pathological vascular remodeling. The current review mainly focuses on endogenous matrix proteins in blood vessels and discusses the interaction of these matrix proteins with other ECM proteins, cell surface receptors, cytokines, complement and coagulation factors, and their potential roles in maintaining vascular homeostasis and preventing pathological remodeling.
Collapse
Affiliation(s)
- Yi Fu
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics (Y.Z.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Kai Wang
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhuofan Li
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|