1
|
Zhang Z, Dong Q, Li Z, Cheng G, Li Z. Bi-phasic integrated silk fibroin/polycaprolactone scaffolds for osteochondral regeneration inspired by the native joint tissue and interface. Mater Today Bio 2025; 32:101737. [PMID: 40275950 PMCID: PMC12018571 DOI: 10.1016/j.mtbio.2025.101737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/05/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
Osteochondral scaffolds designed with bi-phasic and multi-phasic have typically struggled with post-implantation delamination. To address this issue, we developed a novel integrated scaffold with natural and continuous interface and heterogeneous bilayer structure. Through layer-by-layer wet electrospinning, two-dimensional (2D) bi-layer integrated membranes of silk fibroin (SF) and polycaprolactone (PCL) were fabricated. These membranes were then transformed into three-dimensional (3D) scaffolds using a CO2 gas foaming technique, followed by gelatin coating on the osteogenic layer to afford final bi-phasic porous scaffolds. In vitro studies indicated that the 3D scaffolds better-maintained cell phenotypes than conventional 2D electrospun films. Additionally, the 3D scaffolds showed superior cartilage repair and osteoinductivity potential, with increased subchondral bone volume and reduced defect area in rat osteochondral defects models at 12 weeks. Taken together, these gas-foamed scaffolds were a promising candidate for osteochondral regeneration.
Collapse
Affiliation(s)
- Zexing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Qingquan Dong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Zubing Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Gu Cheng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhi Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
2
|
Wang T, Zhao X, Li J, Yin C, Jiang B, Xie J, Wang B, Wang Y, Cao Z, Yao Q, Zheng S, Sui J, Zhu K. Oxytocin-loaded hydrogel promotes cartilage regeneration and regulates microenvironment. Biofabrication 2025; 17:025030. [PMID: 40096748 DOI: 10.1088/1758-5090/adc158] [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: 10/13/2024] [Accepted: 03/17/2025] [Indexed: 03/19/2025]
Abstract
Osteoarthritis is a common orthopedic condition, and traditional treatment methods often fail to regenerate cartilage effectively. Oxytocin (OXT) is a neuropeptide that plays a crucial role in the skeletal system. Hyaluronic acid (HAMA) hydrogel has emerged as a key carrier for cartilage repair due to its excellent biocompatibility and biodegradability. Combining OXT with HAMA hydrogel and implanting it at the site of cartilage defects can effectively promote cartilage regeneration. Cartilage damage often results in an altered microenvironment, characterized by macrophage polarization and high levels of reactive oxygen species (ROS). Oxidative stress can stimulate macrophages to produce more pro-inflammatory factors. OXT can inhibit the secretion of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1βby interacting with the STAT3/NF-κB signaling pathway, as well as the PI3K/Akt and mitogen-activated protein kinase pathways, thereby inducing the polarization of macrophages from the M1 phenotype to the M2 phenotype and alleviating the inflammatory response. OXT can also enhance the expression of NRF and HO-1, which helps eliminate ROS and suppress the expression of pro-inflammatory factors. Regulating the microenvironment of cartilage damage is beneficial for cartilage protection and repair. OXT activates the CFOS/AP-1 and STAT1/JAK2 pathways, which together act on MMP2 and MMP9 to alleviate cartilage degeneration. The STAT1/JAK2 pathway can further increase the expression of Col2, thereby protecting chondrocytes. Additionally, OXT can directly boost the protein levels of SOX9 and COMP, promoting chondrocyte proliferation and cartilage protection, ultimately achieving the therapeutic goal for arthritis. This study explores the potential of HAMA hydrogel as a delivery system for OXT and analyzes their impact on cartilage regeneration and anti-inflammatory properties. This research provides a novel strategy for the treatment of cartilage injuries.
Collapse
Affiliation(s)
- Tianming Wang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Xiao Zhao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Jiayi Li
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Chongchong Yin
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Bo Jiang
- Children's Hospital of Nanjing Medical University, Nanjing 210000, People's Republic of China
| | - Jiaming Xie
- Children's Hospital of Nanjing Medical University, Nanjing 210000, People's Republic of China
| | - Binghao Wang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Yufeng Wang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Zhicheng Cao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Qingqiang Yao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Shengnai Zheng
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Jisheng Sui
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Kun Zhu
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, People's Republic of China
| |
Collapse
|
3
|
Hu L, Liu Q, Wang Y, Wang C, Fan Y, Liu S, Shi Y, Jin K, Tan WQ, Pan P, Chen J. A Semi-Interpenetrating Network Hydrogel with Excellent Photothermal Antibacterial and ROS Scavenging Activities for MRSA-Infected Wounds. ACS APPLIED MATERIALS & INTERFACES 2025; 17:15896-15909. [PMID: 40025822 DOI: 10.1021/acsami.4c17889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
The prolonged infection of bacteria at the wound site may lead to serious physical problems. Herein, a multifunctional macroporous hydrogel with superior photothermal antibacterial and ROS scavenging activity (denoted as M-XG gel) was designed for the treatment of MRSA-infected wounds. The M-XG gels are composed of embedding Prussian blue nanoparticles (PBNPs) as photothermal converters and chelating ferric ions with xanthan gum (XG) and dopamine (DA) to form a semipermeable network. The introduction of DA occupies the cross-link sites of ferric ions, further increasing the pore size (200-500 μm open macropores) and endowing the hydrogel with ideal adhesion. The increase of cross-link sites in PBNPs formed a promising equilibrium M-XG gel with identical macroporous structures and toughened mechanical performance. The metal ligands between ferric ions and catechols, as well as the unique photothermal response of PBNPs, endow the hydrogels with a fast and stable near-infrared (NIR) photothermal conversion efficiency (48%). In the MRSA-infected SD rat trauma model, wounds treated with the M-XG gel group had completely closed after 14 days, effectively controlling wound bacterial infection and accelerating angiogenesis and collagen deposition, synergistically promoting infected wound healing. Therefore, the photothermal hydrogel with a semi-interpenetrating network demonstrates its great potential for infected wound tissue engineering.
Collapse
Affiliation(s)
- Le Hu
- Marine College, Shandong University, Weihai 264209, China
| | - Qing Liu
- Marine College, Shandong University, Weihai 264209, China
| | - Yuxin Wang
- Marine College, Shandong University, Weihai 264209, China
| | - Chunxiao Wang
- Marine College, Shandong University, Weihai 264209, China
| | - Yinuo Fan
- Marine College, Shandong University, Weihai 264209, China
| | - Shuying Liu
- Marine College, Shandong University, Weihai 264209, China
| | - Yujiao Shi
- Marine College, Shandong University, Weihai 264209, China
| | - Kang Jin
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wei-Qiang Tan
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Panpan Pan
- Marine College, Shandong University, Weihai 264209, China
| | - Jingdi Chen
- Marine College, Shandong University, Weihai 264209, China
| |
Collapse
|
4
|
Chen K, Wang F, Sun X, Ge W, Zhang M, Wang L, Zheng H, Zheng S, Tang H, Zhou Z, Wu G. 3D-printed zinc oxide nanoparticles modified barium titanate/hydroxyapatite ultrasound-responsive piezoelectric ceramic composite scaffold for treating infected bone defects. Bioact Mater 2025; 45:479-495. [PMID: 39717367 PMCID: PMC11664295 DOI: 10.1016/j.bioactmat.2024.11.015] [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: 07/25/2024] [Revised: 10/22/2024] [Accepted: 11/10/2024] [Indexed: 12/25/2024] Open
Abstract
Clinically, infectious bone defects represent a significant threat, leading to osteonecrosis, severely compromising patient prognosis, and prolonging hospital stays. Thus, there is an urgent need to develop a bone graft substitute that combines broad-spectrum antibacterial efficacy and bone-inductive properties, providing an effective treatment option for infectious bone defects. In this study, the precision of digital light processing (DLP) 3D printing technology was utilized to construct a scaffold, incorporating zinc oxide nanoparticles (ZnO-NPs) modified barium titanate (BT) with hydroxyapatite (HA), resulting in a piezoelectric ceramic scaffold designed for the repair of infected bone defects. The results indicated that the addition of ZnO-NPs significantly improved the piezoelectric properties of BT, facilitating a higher HA content within the ceramic scaffold system, which is essential for bone regeneration. In vitro antibacterial assessments highlighted the scaffold's potent antibacterial capabilities. Moreover, combining the synergistic effects of low-intensity pulsed ultrasound (LIPUS) and piezoelectricity, results demonstrated that the scaffold promoted notable osteogenic and angiogenic potential, enhancing bone growth and repair. Furthermore, transcriptomics analysis results suggested that the early growth response-1 (EGR1) gene might be crucial in this process. This study introduces a novel method for constructing piezoelectric ceramic scaffolds exhibiting outstanding osteogenic, angiogenic, and antibacterial properties under the combined influence of LIPUS, offering a promising treatment strategy for infectious bone defects.
Collapse
Affiliation(s)
- Kai Chen
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Fang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Xiumei Sun
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Wenwei Ge
- Key Laboratory of Automobile Materials of Ministry of Education, School of Materials Science and Engineering, Jilin University, Changchun, 130021, China
| | - Mingjun Zhang
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Lin Wang
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Haoyu Zheng
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Shikang Zheng
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Haoyu Tang
- Key Laboratory of Automobile Materials of Ministry of Education, School of Materials Science and Engineering, Jilin University, Changchun, 130021, China
| | - Zhengjie Zhou
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Guomin Wu
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| |
Collapse
|
5
|
Han C, Wu Z, Gao Y, Yang S, Wang Y, Guo M, Li Y, Yin W, Liu L, Song W, Zhang P, Wang L. An Additive-Fabricated Biphasic Scaffold for Procedurally Promoting Bone Regeneration via Antioxidant and Osteogenesis. Biotechnol Bioeng 2025; 122:654-666. [PMID: 39682015 DOI: 10.1002/bit.28896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/05/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
The repair process of bone tissue includes the early inflammatory response period and the late tissue repair period. It has been widely approved to be beneficial to the repair of bone injury by procedurally inhibiting the inflammatory response in the early stage and promoting bone regeneration in the late stage. In this study, the nano-hydroxyapatite/Poly(glycolide-co-caprolactone) (n-HA/PGCL) scaffold loaded with icariin was fabricated by fused deposition modeling technique, and the quercetin-loaded GelMA was further filled into the scaffold pores via light-curing methods to form a biphasic scaffold loaded with dual molecules (PHI + GQ scaffold). The releases of icariin and quercetin were sequential due to different degradation rates of GelMA and PGCL. In vitro, the scaffold not only scavenged reactive oxygen species production, but also promoted osteogenic differentiation of the MC-3T3-E1 cells. Furthermore, in vivo bone reconstruction of PHI + GQ scaffold was better than other groups by assessment of micro-CT data. In addition, the immunofluorescence staining of Arg-1 and iNOS indicated that PHI + GQ scaffold created an immune microenvironment conducive to bone repair due to the release of quercetin in the early stage, and HE and Masson staining suggested that PHI + GQ scaffold induced more new bone formation. These results demonstrated that the biphasic scaffold loaded with icariin and quercetin had both antioxidants in the early stage and osteogenesis properties in the late stage, obtaining satisfactory bone repair outcomes. Thus, the biphasic scaffold loaded with icariin and quercetin for sequential release could provide a promising solution for the restoration of bone defects and represent a potential strategy for bone regeneration.
Collapse
Affiliation(s)
- Chunyu Han
- Department of Stomatology, China-Japan Union Hospital, Jilin University, Changchun, P.R. China
| | - Zhenxu Wu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P.R. China
| | - Yuqi Gao
- Department of Stomatology, China-Japan Union Hospital, Jilin University, Changchun, P.R. China
| | - Shuang Yang
- Department of Stomatology, China-Japan Union Hospital, Jilin University, Changchun, P.R. China
| | - Yu Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P.R. China
| | - Min Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P.R. China
| | - Yueyue Li
- Department of Ophthalmology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Wanzhong Yin
- Department of Otorhinolaryngology, First Hospital of Jilin University, Changchun, P.R. China
| | - Ling Liu
- Department of Otorhinolaryngology, First Hospital of Jilin University, Changchun, P.R. China
| | - Wenzhi Song
- Department of Stomatology, China-Japan Union Hospital, Jilin University, Changchun, P.R. China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P.R. China
| | - Liqiang Wang
- Department of Ophthalmology, Chinese PLA General Hospital, Beijing, P.R. China
| |
Collapse
|
6
|
Klimek K, Terpilowska S, Michalak A, Bernacki R, Nurzynska A, Cucchiarini M, Tarczynska M, Gaweda K, Głuszek S, Ginalska G. Modern Approach to Testing the Biocompatibility of Osteochondral Scaffolds in Accordance with the 3Rs Principle─Preclinical In Vitro, Ex Vivo, and In Vivo Studies Using the Biphasic Curdlan-Based Biomaterial. ACS Biomater Sci Eng 2025; 11:845-865. [PMID: 39832791 DOI: 10.1021/acsbiomaterials.4c01107] [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: 01/22/2025]
Abstract
The aim of this work is to provide a comprehensive set of biological tests to assess the biomedical potential of novel osteochondral scaffolds with methods proposed to comply with the 3Rs principle, focusing here on a biphasic Curdlan-based osteochondral scaffold as a promising model biomaterial. In vitro experiments include the evaluation of cytotoxicity, mutagenicity, and genotoxicity referring to ISO standards, the assessment of the viability and proliferation of human chondrocytes and osteoblasts, and the estimation of inflammation after direct contact of biomaterials with human macrophages. Ex vivo experiments include assessments of the response of the surrounding osteochondral tissue after incubation with the implanted biomaterial. In vivo experiments involve an evaluation of the toxicity and regenerative potential of the biomaterial in zebrafish (larvae and adults) and in osteochondral defects in dogs (veterinary patients). The applied set of tests allows us to show that the Curdlan-based scaffold does not induce cytotoxicity (cell viability close to 100%), mutagenicity (the level of reversion is not 2× higher compared to the control), and genotoxicity (it does not exhibit any change in chromosomal aberration; the frequency of micronuclei, micronucleated binucleated cells, and cytokinesis-block proliferation index is comparable to the control; moreover, it does not cause the formation of comets in cells). This biomaterial also promotes the viability and proliferation of chondrocytes and osteoblasts (the OD values between the fourth and seventh day of incubation increase by approximately 1.6×). The Curdlan-based scaffold stimulates only a transient inflammatory response in vitro and ex vivo. This biomaterial does not cause Danio rerio larvae malformation and also enables proper regeneration of the caudal fin in adults. Finally, it supports the regeneration of an osteochondral defect in veterinary patients. Thus, this is a proposal to use alternative methods for biological assessment of osteochondral scaffolds as opposed to commonly used tests using large numbers of laboratory animals.
Collapse
Affiliation(s)
- Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
| | - Sylwia Terpilowska
- Department of Surgical Medicine with the Laboratory of Medical Genetics, Jan Kochanowski University, Collegium Medicum, IX Wiekow Kielc 19A Av., 25-317 Kielce, Poland
| | - Agnieszka Michalak
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, Chodzki 4a Street, 20-093 Lublin, Poland
| | - Rafal Bernacki
- Veterinary Clinic Aura, Debowa 31 Street, 86-065 Lochowo, Poland
| | - Aleksandra Nurzynska
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Saarland University, Kirrbergerstr. Bldg 37, 66421 Homburg/Saar, Germany
| | - Marta Tarczynska
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-954 Lublin, Poland
| | - Krzysztof Gaweda
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-954 Lublin, Poland
| | - Stanisław Głuszek
- Department of Surgical Medicine with the Laboratory of Medical Genetics, Jan Kochanowski University, Collegium Medicum, IX Wiekow Kielc 19A Av., 25-317 Kielce, Poland
| | - Grazyna Ginalska
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
- Faculty of Health Sciences, Vincent Pol University, Choiny 2 Street, 20-816 Lublin, Poland
| |
Collapse
|
7
|
Shen C, Zhou Z, Li R, Yang S, Zhou D, Zhou F, Geng Z, Su J. Silk fibroin-based hydrogels for cartilage organoids in osteoarthritis treatment. Theranostics 2025; 15:560-584. [PMID: 39744693 PMCID: PMC11671376 DOI: 10.7150/thno.103491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/06/2024] [Indexed: 01/11/2025] Open
Abstract
Osteoarthritis (OA) is a common joint disease characterized by cartilage degeneration. It can cause severe pain, deformity and even amputation risk. However, existing clinical treatment methods for cartilage repair present certain deficiencies. Meanwhile, the repair effect of cartilage tissue engineering is also unsatisfactory. Cartilage organoids are multicellular aggregates with cartilage-like three-dimensional structure and function. On the one hand, cartilage organoids can be used to explore the pathogenesis of OA by constructing disease models. On the other hand, it can be used as filler for rapid cartilage repair. Extracellular matrix (ECM)-like three-dimensional environment is the key to construct cartilage organoids. Silk fibroin (SF)-based hydrogels not only have ECM-like structure, but also have unique mechanical properties and remarkable biocompatibility. Therefore, SF-based hydrogels are considered as ideal biomaterials for constructing cartilage organoids. In this review, we reviewed the studies of cartilage organoids and SF-based hydrogels. The advantages of SF-based hydrogels in constructing cartilage organoids and the iterative optimization of cartilage organoids through designing hydrogels by using artificial intelligence (AI) calculation are also discussed. This review aims to provide a theoretical basis for the treatment of OA using SF-based biomaterials and cartilage organoids.
Collapse
Affiliation(s)
- Congyi Shen
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Ziyang Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Ruiyang Li
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shike Yang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Anesthesiology, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Dongyang Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
8
|
Lun W, Wang H, Li M, Ma J, Ding Y, Zheng X, Cao X, Li Q. Fabrication of MnO 2-Modified Decellularized Tendon Membrane for Enhancing Tendon Repair. Adv Healthc Mater 2025; 14:e2402584. [PMID: 39491818 DOI: 10.1002/adhm.202402584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/11/2024] [Indexed: 11/05/2024]
Abstract
Repairing tendon/ligament injuries is a major challenge in sports medicine. It has been reported that tendon injury healing is hindered by massive production of reactive oxygen species (ROS). Manganese oxides nanoparticles are generally non-toxic, can scavenge ROS, promote tissue regeneration, and hold promise for sustainable nanotechnologies. However, the effective and safe integration of MnO2 nanoparticles on decellularized scaffold mediating tissue repair is still a great challenge. To address these issues, an in situ MnO2-modified decellularized scaffold is developed to enhance tendon regeneration through improving microenvironment. The decellularized fibrous membrane is designed and prepared using the central tendon of the porcine diaphragm. Then MnO2 nanozymes are in situ grown on the collagen fibers using tannic acid (TA) as cross-linking agent and reducing agent. The results showed that MnO2-modified scaffold eliminates excessive accumulation of ROS in cells, protects mitochondrial, and maintains the phenotype of tendon cells in an oxidative stress environment. Notably, it is found that the MnO2-modified scaffold exhibits good biocompatibility and is able to promote the tendon healing in the rat patellar tendon defect model. Altogether, this study confirmed that this nanozyme-functionalized decellularized extracellular matrix effectively enhanced tendon repair by scavenging ROS, which provides new strategies for enhancing tendon regeneration.
Collapse
Affiliation(s)
- Wanqing Lun
- School of Medicine, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, P. R. China
| | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, P. R. China
- The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, 510630, P. R. China
| | - Mengyuan Li
- Division of Joint Osteopathy and Traumatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, P. R. China
| | - Jiuzhi Ma
- School of Medicine, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, P. R. China
| | - Yilin Ding
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, P. R. China
- Department of Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, P. R. China
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, P. R. China
- The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, 510630, P. R. China
| | - Xiaodong Cao
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, P. R. China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Qingtao Li
- School of Medicine, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, P. R. China
| |
Collapse
|
9
|
Yang J, Zhang L, Wang Y, Wang N, Wei H, Zhang S, Ding Q, Sun S, Ding C, Liu W. Dihydromyricetin-loaded oxidized polysaccharide/L-arginine chitosan adhesive hydrogel promotes bone regeneration by regulating PI3K/AKT signaling pathway and MAPK signaling pathway. Carbohydr Polym 2024; 346:122614. [PMID: 39245525 DOI: 10.1016/j.carbpol.2024.122614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024]
Abstract
Bone defects caused by trauma, infection and congenital diseases still face great challenges. Dihydromyricetin (DHM) is a kind of flavone extracted from Ampelopsis grossedentata, a traditional Chinese medicine. DHM can enhance the osteogenic differentiation of human bone marrow mesenchymal stem cells with the potential to promote bone regeneration. Hydrogel can be used as a carrier of DHM to promote bone regeneration due to its unique biochemical characteristics and three-dimensional structure. In this study, oxidized phellinus igniarius polysaccharides (OP) and L-arginine chitosan (CA) are used to develop hydrogel. The pore size and gel strength of the hydrogel can be changed by adjusting the oxidation degree of oxidized phellinus igniarius polysaccharides. The addition of DHM further reduce the pore size of the hydrogel (213 μm), increase the mechanical properties of the hydrogel, and increase the antioxidant and antibacterial activities of the hydrogel. The scavenging rate of DPPH are 72.30 ± 0.33 %, and the inhibition rate of E.coli and S.aureus are 93.12 ± 0.38 % and 94.49 ± 1.57 %, respectively. In addition, PCAD has good adhesion and biocompatibility, and its extract can effectively promote the osteogenic differentiation of MC3T3-E1 cells. Network pharmacology and molecular docking show that the promoting effect of DHM on osteogenesis may be achieved by activating the PI3K/AKT and MAPK signaling pathways. This is confirmed through in vitro cell experiments and in vivo animal experiments.
Collapse
Affiliation(s)
- Jiali Yang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Lifeng Zhang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Yue Wang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Ning Wang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Hewei Wei
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Shuai Zhang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Qiteng Ding
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Shuwen Sun
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Chuanbo Ding
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China.
| | - Wencong Liu
- School of Food and Pharmaceutical Engineering, Wuzhou University, Wuzhou 543002, China.
| |
Collapse
|
10
|
Yang Y, Yu M, Mo Y, Cheng Y, Huang B, Wang W, Zhu M, Jia X, Feng L, Yang B. Metal-ion-binding properties of glycyrrhiza polysaccharide extracted from Licorice: Structural characterization and potential application in drug delivery. Carbohydr Polym 2024; 346:122658. [PMID: 39245514 DOI: 10.1016/j.carbpol.2024.122658] [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: 05/06/2024] [Revised: 07/27/2024] [Accepted: 08/24/2024] [Indexed: 09/10/2024]
Abstract
Licorice is not only a widely used food, but also a classic tonic Chinese medicine, which mainly contains glycyrrhiza polysaccharides (GP) and flavonoids with excellent anti-inflammatory and antioxidant pharmacological activities. In this study, a neutral homogeneous polysaccharide (GP1-2) was isolated from Glycyrrhiza uralensis Fisch. However, its gelation behavior and properties have yet to be comprehensively studied. In this study, a Ca2+ cross-linked physical hydrogel based on neutral GP1-2 (GP1-2-Ca2+) is fabricated. The ability of metal ions to cross-linked gelation with GP1-2 is explored with respect to the polysaccharide concentrations, ion species, and pH environments. The pH range of Ca2+ cross-linked with GP1-2 to form hydrogel is 8 to 10, and the gelation concentration ranges from 20.0 % to 50.0 % w/v. Subsequently, the properties of the GP1-2-Ca2+ hydrogels are investigated using rheological measurements, scanning electron microscopy, free radical scavenging, MTT assays, healing capability, and enzyme-linked immunosorbent assays. The results reveal that the structure of GP1-2 presents an irregular porous structure, however, the physical gel formed after cross-linking with Ca2+ microscopically showed a globular porous structure with uniform distribution, suggesting that this structure characteristic may be used as a carrier material for drug delivery. Meanwhile, the GP1-2-Ca2+ hydrogel also possessed extraordinary viscoelasticity, cytocompatibility, antioxidant properties, anti-inflammatory activity, and ability to promote wound healing. Furthermore, the potential of GP1-2-Ca2+ hydrogels as drug delivery materials was validated by using rhein as a model drug for encapsulation, it is demonstrated that its cumulative release behavior of GP1-2-Ca2+ is pH-dependent. All in all, this study reveals the potential application of natural polysaccharides in drug delivery, highlighting its dual roles as carrier materials and bioactive ingredients.
Collapse
Affiliation(s)
- Yanjun Yang
- School of Traditional Chinese Pharmacy, Jiangning Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, PR China
| | - Mengli Yu
- School of Traditional Chinese Pharmacy, Jiangning Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yulin Mo
- School of Traditional Chinese Pharmacy, Jiangning Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yue Cheng
- School of Traditional Chinese Pharmacy, Jiangning Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, PR China
| | - Bin Huang
- School of Traditional Chinese Pharmacy, Jiangning Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, PR China
| | - Weilin Wang
- School of Traditional Chinese Pharmacy, Jiangning Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, PR China
| | - Maomao Zhu
- School of Traditional Chinese Pharmacy, Jiangning Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, Jiangning Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Liang Feng
- School of Traditional Chinese Pharmacy, Jiangning Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Bing Yang
- School of Traditional Chinese Pharmacy, Jiangning Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
11
|
Cui X, Huang C, Huang Y, Zhang Y, Wu J, Wang G, Zhou XZ, Zhang J, Wang L, Cheng L, Zhang KQ. Amplification of Metalloregulatory Proteins in Macrophages by Bioactive ZnMn@SF Hydrogels for Spinal Cord Injury Repair. ACS NANO 2024; 18:33614-33628. [PMID: 39579147 DOI: 10.1021/acsnano.4c12236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Macrophages are rapidly activated and polarized toward the M1 phenotype after spinal cord injury (SCI), and inhibiting M1-like macrophages has emerged as a promising SCI treatment approach. Metalloregulatory proteins, which sense specific metal ions with high affinity and specificity, play a critical role in immune regulation. Here, we screened various bioactive metal ions associated with metalloregulatory proteins and discovered that Zn2+ and Mn2+ effectively suppressed M1 polarization. Based on these findings, mildly alkaline ZnMn-based layered double hydroxides (ZnMn-LDHs) self-assembled from Zn2+ coordinated with Mn2+ were developed to inhibit M1-like macrophages. ZnMn-LDHs effectively neutralized the acidic environment and promoted the expression of metalloregulatory proteins, including metallothionein (MT), superoxide dismutase 1 (SOD1), and superoxide dismutase 2 (SOD2), thereby eliciting robust M1-like macrophage inhibition. More importantly, nerve growth factor (NGF) released by macrophages following the regulation by ZnMn-LDHs promoted the elongation and spreading of Schwann cells. By integrating ZnMn-LDHs with silk fibroin (SF), ZnMn@SF injectable hydrogels were constructed for SCI repair. An in vivo animal model further revealed the excellent anti-inflammatory effects of the ZnMn@SF hydrogels in treating SCI, which promoted functional recovery. Our findings underscore the importance of metalloregulatory proteins regulated by metal ions in inhibiting M1-like macrophages, providing a promising therapeutic strategy for SCI treatment.
Collapse
Affiliation(s)
- Xiaoliang Cui
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Cheng Huang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Yechen Huang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Yuxuan Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Jie Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Gang Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Xiao-Zhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Jun Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Li Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Ke-Qin Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| |
Collapse
|
12
|
Chen Y, Xu W, Pan Z, Li B, Mo X, Li Y, Wang J, Wang Y, Wei Z, Chen Y, Han Z, Lin C, Liu Y, Ye X, Yu J. Three-dimensional gas-foamed scaffolds decorated with metal phenolic networks for cartilage regeneration. Mater Today Bio 2024; 29:101249. [PMID: 39351488 PMCID: PMC11440796 DOI: 10.1016/j.mtbio.2024.101249] [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: 05/20/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
Inflammation is a major impediment to the healing of cartilage injuries, yet bioactive scaffolds suitable for cartilage repair in inflammatory environments are extremely rare. Herein, we utilized electrospinning to fabricate a two-dimensional nanofiber scaffold (2DS), which was then subjected to gas foaming to obtain a three-dimensional scaffold (3DS). 3DS was modified with metal phenolic networks (MPNs) composed of epigallocatechin gallate (EGCG) and strontium ions (Sr2+) to afford a MPNs-modified 3D scaffold (3DS-E). Gas-foamed scaffold exhibited multilayered structure conducive to cellular infiltration and proliferation. Compared to other groups, 3DS-E better preserved chondrocytes under interleukin (IL)-1β induced inflammatory environment, showing less apoptosis of chondrocytes and higher expression of cartilage matrix. Additionally, 3DS-E facilitated the regeneration of more mature cartilage in vivo, reduced cell apoptosis, and decreased the expression of pro-inflammatory cytokines. Taken together, 3DS-E may offer an ideal candidate for cartilage regeneration.
Collapse
Affiliation(s)
- Yujie Chen
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Wei Xu
- Department of Plastic Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, 266035, China
| | - Zhen Pan
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Bohui Li
- Plastic Surgery Institute, Shandong Second Medical University, Weifang, Shandong, 261053, China
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Huangpu, Shanghai, 200001, China
| | - Xiumei Mo
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Songjiang, Shanghai, 201600, China
| | - Yucai Li
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Jielin Wang
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Yuan Wang
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Zhenyuan Wei
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Yicheng Chen
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Zhaopu Han
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Chen Lin
- Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, Pudong New Area, Shanghai, 200120, China
| | - Yu Liu
- Plastic Surgery Institute, Shandong Second Medical University, Weifang, Shandong, 261053, China
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Huangpu, Shanghai, 200001, China
| | - Xiaojian Ye
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Jiangming Yu
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, 200336, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| |
Collapse
|
13
|
Ilic-Tomic T, Kramar A, Kostic M, Vojnovic S, Milovanovic J, Petkovic M, D’Agostino PM, Gulder TAM, Nikodinovic-Runic J. Functionalization of silk with actinomycins from Streptomyces anulatus BV365 for biomedical applications. Front Bioeng Biotechnol 2024; 12:1466757. [PMID: 39364265 PMCID: PMC11447452 DOI: 10.3389/fbioe.2024.1466757] [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: 07/18/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024] Open
Abstract
Silk, traditionally acclaimed as the "queen of fiber," has been widely used thanks to its brilliant performance such as gentleness, smoothness and comfortableness. Owing to its mechanical characteristics and biocompatibility silk has a definitive role in biomedical applications, both as fibroin and fabric. In this work, the simultaneous dyeing and functionalization of silk fabric with pigments from Streptomyces anulatus BV365 were investigated. This strain produced high amounts of orange extracellular pigments on mannitol-soy flour agar, identified as actinomycin D, C2 and C3. The application of purified actinomycins in the dyeing of multifiber fabric was assessed. Actinomycins exhibited a high affinity towards protein fibers (silk and wool), but washing durability was maintained only with silk. Acidic condition (pH5) and high temperature (65°C) facilitated the silk dyeing. The morphologies and chemical components of the treated silk fabrics were analyzed using scanning electron microscopy and Fourier transform infrared spectroscopy. The results showed the pigments bind to the silk through interaction with the carbonyl group in silk fibroin rendering the functionalized, yet surface that does not cause skin irritation. The treated silk exhibited a remarkable antibacterial effect, while the biocompatibility test performed with 3D-reconstructed human epidermis model indicated safe biological properties, paving the way for future application of this material in medicine.
Collapse
Affiliation(s)
- Tatjana Ilic-Tomic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Ana Kramar
- Department of Textile Engineering, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
- Novel Materials and Nanotechnology Group, Institute of Agrochemistry and Food Technology (IATA), Spanish Council for Scientific Research (CSIC), Paterna, Spain
| | - Mirjana Kostic
- Department of Textile Engineering, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Sandra Vojnovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jelena Milovanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Milos Petkovic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Paul M. D’Agostino
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Saarland, Germany
- Technical University of Dresden, Dresden, Saxony, Germany
| | - Tobias A. M. Gulder
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Saarland, Germany
- Technical University of Dresden, Dresden, Saxony, Germany
| | | |
Collapse
|
14
|
Liu Y, Zhang H, Chen T, Xu C, Bao X. Metal-organic frameworks (MOFs) and their derivatives as emerging biomaterials for the treatment of osteoarthritis. Front Pharmacol 2024; 15:1462368. [PMID: 39359247 PMCID: PMC11444981 DOI: 10.3389/fphar.2024.1462368] [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: 07/10/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
As a novel class of smart biomaterials with promising potentials, metal-organic frameworks (MOFs) are widely utilized in the field of biomedicine. Current researches indicate that the therapeutic strategies for osteoarthritis (OA) are highly limited to achieving symptom improvement and reducing both pain and inflammation. Together, the introduction of MOFs into the treatment of OA holds the potential to offer significant benefits. This is because MOFs not only have intrinsic biological activities, but also act as carriers to facilitate controlled drug delivery and prolong the duration in the management of OA. This paper presents a review of the recent studies that have explored the potential usage of MOFs as drugs or carriers in the treatment of OA, which also examines the progress of MOFs in tissue engineering for the treatment of OA. These studies are anticipated to not only enhance the comprehension of MOFs but also provide strong evidence in favor of their utilization in the treatment of OA.
Collapse
Affiliation(s)
- Yufu Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Hongwei Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Tianyan Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Chang Xu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Xingfu Bao
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
15
|
Yu Q, Xiao Y, Guan M, Zhang X, Yu J, Han M, Li Z. Copper metabolism in osteoarthritis and its relation to oxidative stress and ferroptosis in chondrocytes. Front Mol Biosci 2024; 11:1472492. [PMID: 39329090 PMCID: PMC11425083 DOI: 10.3389/fmolb.2024.1472492] [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: 07/29/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Ferroptosis, an iron-ion-dependent process of lipid peroxidation, damages the plasma membrane, leading to non-programmed cell death. Osteoarthritis (OA), a prevalent chronic degenerative joint disease among middle-aged and older adults, is characterized by chondrocyte damage or loss. Emerging evidence indicates that chondrocyte ferroptosis plays a role in OA development. However, most research has concentrated on ferroptosis regulation involving typical iron ions, potentially neglecting the significance of elevated copper ions in both serum and joint fluid of patients with OA. This review aims to fill this gap by systematically examining the interplay between copper metabolism, oxidative stress, ferroptosis, and copper-associated cell death in OA. It will provide a comprehensive overview of copper ions' role in regulating ferroptosis and their dual role in OA. This approach seeks to offer new insights for further research, prevention, and treatment of OA.
Collapse
Affiliation(s)
- Qingyuan Yu
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Yanan Xiao
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Mengqi Guan
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xianshuai Zhang
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jianan Yu
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Mingze Han
- Clinical College of Integrated Traditional Chinese and Western Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Zhenhua Li
- Orthopedic Center, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| |
Collapse
|
16
|
Younus ZM, Ahmed I, Roach P, Forsyth NR. A phosphate glass reinforced composite acrylamide gradient scaffold for osteochondral interface regeneration. BIOMATERIALS AND BIOSYSTEMS 2024; 15:100099. [PMID: 39221155 PMCID: PMC11364006 DOI: 10.1016/j.bbiosy.2024.100099] [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: 02/27/2024] [Revised: 07/12/2024] [Accepted: 07/20/2024] [Indexed: 09/04/2024] Open
Abstract
The bone-cartilage interface is defined by a unique arrangement of cells and tissue matrix. Injury to the interface can contribute to the development of arthritic joint disease. Attempts to repair osteochondral damage through clinical trials have generated mixed outcomes. Tissue engineering offers the potential of integrated scaffold design with multiregional architecture to assist in tissue regeneration, such as the bone-cartilage interface. Challenges remain in joining distinct materials in a single scaffold mass while maintaining integrity and avoiding delamination. The aim of the current work is to examine the possibility of joining two closely related acrylamide derivatives such as, poly n-isopropyl acrylamide (pNIPAM) and poly n‑tert‑butyl acrylamide (pNTBAM). The target is to produce a single scaffold unit with distinct architectural regions in the favour of regenerating the osteochondral interface. Longitudinal phosphate glass fibres (PGFs) with the formula 50P2O5.30CaO.20Na2O were incorporated to provide additional bioactivity by degradation to release ions such as calcium and phosphate which are considered valuable to assist the mineralization process. Polymers were prepared via atom transfer radical polymerization (ATRP) and solutions cast to ensure the integration of polymers chains. Scaffold was characterized using scanning electron microscope (SEM) and Fourier transform infra-red (FTIR) techniques. The PGF mass degradation pattern was inspected using micro computed tomography (µCT). Biological assessment of primary human osteoblasts (hOBs) and primary human chondrocytes (hCHs) upon scaffolds was performed using alizarin red and colorimetric calcium assay for mineralization assessment; alcian blue staining and dimethyl-methylene blue (DMMB) assay for glycosaminoglycans (GAGs); immunostaining and enzyme-linked immunosorbent assay (ELISA) to detect functional proteins expression by cells such as collagen I, II, and annexin A2. FTIR analysis revealed an intact unit with gradual transformation from pNIPAM to pNTBAM. SEM images showed three distinct architectural regions with mean pore diameter of 54.5 µm (pNIPAM), 16.5 µm (pNTBAM) and 118 µm at the mixed interface. Osteogenic and mineralization potential by cells was observed upon the entire scaffold's regions. Chondrogenic activity was relevant on the pNTBAM side of the scaffold only with minimal evidence in the pNIPAM region. PGFs increased mineralization potential of both hOBs and hCHs, evidenced by elevated collagens I, X, and annexin A2 with reduction of collagen II in PGFs scaffolds. In conclusion, pNIPAM and pNTBAM integration created a multiregional scaffold with distinct architectural regions. Differential chondrogenic, osteogenic, and mineralized cell performance, in addition to the impact of PGF, suggests a potential role for phosphate glass-incorporated, acrylamide-derivative scaffolds in osteochondral interface regeneration.
Collapse
Affiliation(s)
- Zaid M. Younus
- School of Pharmacy and Bioengineering, Keele University, Keele, UK
- College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Ifty Ahmed
- Faculty of Engineering, Advanced Materials Research Group, University of Nottingham, Nottingham, UK
| | - Paul Roach
- Department of Chemistry, School of Science, Loughborough University, Leicestershire, UK
| | - Nicholas R. Forsyth
- School of Pharmacy and Bioengineering, Keele University, Keele, UK
- Vice principals’ office, King's College, University of Aberdeen, Aberdeen, AB24 3FX, UK
| |
Collapse
|
17
|
Hu C, Huang R, Xia J, Hu X, Xie D, Jin Y, Qi W, Zhao C, Hu Z. A nanozyme-functionalized bilayer hydrogel scaffold for modulating the inflammatory microenvironment to promote osteochondral regeneration. J Nanobiotechnology 2024; 22:445. [PMID: 39069607 PMCID: PMC11283693 DOI: 10.1186/s12951-024-02723-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: 01/12/2024] [Accepted: 07/14/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND The incidence of osteochondral defects caused by trauma, arthritis or tumours is increasing annually, but progress has not been made in terms of treatment methods. Due to the heterogeneous structure and biological characteristics of cartilage and subchondral bone, the integration of osteochondral repair is still a challenge. RESULTS In the present study, a novel bilayer hydrogel scaffold was designed based on anatomical characteristics to imitate superficial cartilage and subchondral bone. The scaffold showed favourable biocompatibility, and the addition of an antioxidant nanozyme (LiMn2O4) promoted reactive oxygen species (ROS) scavenging by upregulating antioxidant proteins. The cartilage layer effectively protects against chondrocyte degradation in the inflammatory microenvironment. Subchondral bionic hydrogel scaffolds promote osteogenic differentiation of rat bone marrow mesenchymal stem cells (BMSCs) by regulating the AMPK pathway in vitro. Finally, an in vivo rat preclinical osteochondral defect model confirmed that the bilayer hydrogel scaffold efficiently promoted cartilage and subchondral bone regeneration. CONCLUSIONS In general, our biomimetic hydrogel scaffold with the ability to regulate the inflammatory microenvironment can effectively repair osteochondral defects. This strategy provides a promising method for regenerating tissues with heterogeneous structures and biological characteristics.
Collapse
Affiliation(s)
- Chuan Hu
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Ruipeng Huang
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Jiechao Xia
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Xianjing Hu
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhu, 325000, China
| | - Dingqi Xie
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Yang Jin
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Weiming Qi
- Zhejiang Center for Medical Device Evaluation, Zhejiang Medical Products Administration, Hangzhou, 310009, China.
| | - Chengliang Zhao
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Zhijun Hu
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
18
|
Novotná R, Franková J. Materials Suitable for Osteochondral Regeneration. ACS OMEGA 2024; 9:30097-30108. [PMID: 39035913 PMCID: PMC11256084 DOI: 10.1021/acsomega.4c04789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024]
Abstract
Osteochondral defects affect articular cartilage, calcified cartilage, and subchondral bone. The main problem that they cause is a different behavior of cell tissue in the osteochondral and bone part. Articular cartilage is composed mainly of collagen II, glycosaminoglycan (GAG), and water, and has a low healing ability due to a lack of vascularization. However, bone tissue is composed of collagen I, proteoglycans, and inorganic composites such as hydroxyapatite. Due to the discrepancy between the characters of these two parts, it is difficult to find materials that will meet all the structural and other requirements for effective regeneration. When designing a scaffold for an osteochondral defect, a variety of materials are available, e.g., polymers (synthetic and natural), inorganic particles, and extracellular matrix (ECM) components. All of them require the accurate characterization of the prepared materials and a number of in vitro and in vivo tests before they are applied to patients. Taken in concert, the final material needs to mimic the structural, morphological, chemical, and cellular demands of the native tissue. In this review, we present an overview of the structure and composition of the osteochondral part, especially synthetic materials with additives appropriate for healing osteochondral defects. Finally, we summarize in vitro and in vivo methods suitable for evaluating materials for restoring osteochondral defects.
Collapse
Affiliation(s)
- Renáta Novotná
- Department
of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, Olomouc 775 15, Czech Republic
| | - Jana Franková
- Department
of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, Olomouc 775 15, Czech Republic
| |
Collapse
|
19
|
Zheng W, Meng Z, Zhu Z, Wang X, Xu X, Zhang Y, Luo Y, Liu Y, Pei X. Metal-Organic Framework-Based Nanomaterials for Regulation of the Osteogenic Microenvironment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310622. [PMID: 38377299 DOI: 10.1002/smll.202310622] [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: 11/19/2023] [Revised: 02/01/2024] [Indexed: 02/22/2024]
Abstract
As the global population ages, bone diseases have become increasingly prevalent in clinical settings. These conditions often involve detrimental factors such as infection, inflammation, and oxidative stress that disrupt bone homeostasis. Addressing these disorders requires exogenous strategies to regulate the osteogenic microenvironment (OME). The exogenous regulation of OME can be divided into four processes: induction, modulation, protection, and support, each serving a specific purpose. To this end, metal-organic frameworks (MOFs) are an emerging focus in nanomedicine, which show tremendous potential due to their superior delivery capability. MOFs play numerous roles in OME regulation such as metal ion donors, drug carriers, nanozymes, and photosensitizers, which have been extensively explored in recent studies. This review presents a comprehensive introduction to the exogenous regulation of OME by MOF-based nanomaterials. By discussing various functional MOF composites, this work aims to inspire and guide the creation of sophisticated and efficient nanomaterials for bone disease management.
Collapse
Affiliation(s)
- Wenzhuo Zheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zihan Meng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhou Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xu Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiangrui Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yaowen Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yankun Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yanhua Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
20
|
Zhang M, Wang H, Dai GC, Lu PP, Gao YC, Cao MM, Li YJ, Rui YF. Injectable self-assembled GDF5-containing dipeptide hydrogels for enhanced tendon repair. Mater Today Bio 2024; 26:101046. [PMID: 38600922 PMCID: PMC11004210 DOI: 10.1016/j.mtbio.2024.101046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
Owing to the tissue characteristics of tendons with few blood vessels and cells, the regeneration and repair of injured tendons can present a considerable challenge, which considerably affects the motor function of limbs and leads to serious physical and mental pain, along with an economic burden on patients. Herein, we designed and fabricated a dipeptide hydrogel (DPH) using polypeptides P11-4 and P11-8. This hydrogel exhibited self-assembly characteristics and could be administered in vitro. To endow the hydrogel with differentiation and regeneration abilities, we added different concentrations of growth differentiation factor 5 (GDF5) to form GDF5@DPH. GDF5@DPH promoted the aggregation and differentiation of tendon stem/progenitor cells and promoted the regeneration and repair of tendon cells and collagen fibers in injured areas. In addition, GDF5@DPH inhibited inflammatory reactions in the injured area. Owing to its injectable properties, DPH can jointly inhibit adhesion and scar hyperplasia between tissues caused by endogenous inflammation and exogenous surgery and can provide a favorable internal environment for the regeneration and repair of the injured area. Overall, the GDF5@DPH system exhibits considerable promise as a novel approach to treating tendon injury.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Hao Wang
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Guan-Chun Dai
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Pan-Pan Lu
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Yu-Cheng Gao
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Mu-Ming Cao
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Ying-Juan Li
- Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Yun-Feng Rui
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| |
Collapse
|
21
|
Stevanović M, Filipović N. A Review of Recent Developments in Biopolymer Nano-Based Drug Delivery Systems with Antioxidative Properties: Insights into the Last Five Years. Pharmaceutics 2024; 16:670. [PMID: 38794332 PMCID: PMC11125366 DOI: 10.3390/pharmaceutics16050670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
In recent years, biopolymer-based nano-drug delivery systems with antioxidative properties have gained significant attention in the field of pharmaceutical research. These systems offer promising strategies for targeted and controlled drug delivery while also providing antioxidant effects that can mitigate oxidative stress-related diseases. Generally, the healthcare landscape is constantly evolving, necessitating the continual development of innovative therapeutic approaches and drug delivery systems (DDSs). DDSs play a pivotal role in enhancing treatment efficacy, minimizing adverse effects, and optimizing patient compliance. Among these, nanotechnology-driven delivery approaches have garnered significant attention due to their unique properties, such as improved solubility, controlled release, and targeted delivery. Nanomaterials, including nanoparticles, nanocapsules, nanotubes, etc., offer versatile platforms for drug delivery and tissue engineering applications. Additionally, biopolymer-based DDSs hold immense promise, leveraging natural or synthetic biopolymers to encapsulate drugs and enable targeted and controlled release. These systems offer numerous advantages, including biocompatibility, biodegradability, and low immunogenicity. The utilization of polysaccharides, polynucleotides, proteins, and polyesters as biopolymer matrices further enhances the versatility and applicability of DDSs. Moreover, substances with antioxidative properties have emerged as key players in combating oxidative stress-related diseases, offering protection against cellular damage and chronic illnesses. The development of biopolymer-based nanoformulations with antioxidative properties represents a burgeoning research area, with a substantial increase in publications in recent years. This review provides a comprehensive overview of the recent developments within this area over the past five years. It discusses various biopolymer materials, fabrication techniques, stabilizers, factors influencing degradation, and drug release. Additionally, it highlights emerging trends, challenges, and prospects in this rapidly evolving field.
Collapse
Affiliation(s)
- Magdalena Stevanović
- Group for Biomedical Engineering and Nanobiotechnology, Institute of Technical Sciences of SASA, Kneza Mihaila 35/IV, 11000 Belgrade, Serbia;
| | | |
Collapse
|
22
|
Shu C, Qin C, Wu A, Wang Y, Zhao C, Shi Z, Niu H, Chen J, Huang J, Zhang X, Huan Z, Chen L, Zhu M, Zhu Y. 3D Printing of Cobalt-Incorporated Chloroapatite Bioceramic Composite Scaffolds with Antioxidative Activity for Enhanced Osteochondral Regeneration. Adv Healthc Mater 2024; 13:e2303217. [PMID: 38363057 DOI: 10.1002/adhm.202303217] [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: 09/22/2023] [Revised: 02/05/2024] [Indexed: 02/17/2024]
Abstract
Osteochondral defects are often accompanied by excessive reactive oxygen species (ROS) caused by osteoarthritis or acute surgical inflammation. An inflammatory environment containing excess ROS will not only hinder tissue regeneration but also impact the quality of newly formed tissues. Therefore, there is an urgent need to develop scaffolds with both ROS scavenging and osteochondral repair functions to promote and protect osteochondral tissue regeneration. In this work, by using 3D printing technology, a composite scaffold based on cobalt-incorporated chloroapatite (Co-ClAP) bioceramics, which possesses ROS-scavenging activity and can support cell proliferation, adhesion, and differentiation, is developed. Benefiting from the catalytic activity of Co-ClAP bioceramics, the composite scaffold can protect cells from oxidative damage under ROS-excessive conditions, support their directional differentiation, and simultaneously mediate an anti-inflammatory microenvironment. In addition, it is also confirmed by using rabbit osteochondral defect model that the Co-ClAP/poly(lactic-co-glycolic acid) scaffold can effectively promote the integrated regeneration of cartilage and subchondral bone, exhibiting an ideal repair effect in vivo. This study provides a promising strategy for the treatment of defects with excess ROS and inflammatory microenvironments.
Collapse
Affiliation(s)
- Chaoqin Shu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Chen Qin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Aijun Wu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Yufeng Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Chaoqian Zhao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Zhe Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Huicong Niu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 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, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jimin Huang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xinxin Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhiguang Huan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Lei Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Min Zhu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Yufang Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
23
|
Singh A, Sharma JJ, Mohanta B, Sood A, Han SS, Sharma A. Synthetic and biopolymers-based antimicrobial hybrid hydrogels: a focused review. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:675-716. [PMID: 37943320 DOI: 10.1080/09205063.2023.2278814] [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: 08/15/2023] [Accepted: 10/29/2023] [Indexed: 11/10/2023]
Abstract
The constantly accelerating occurrence of microbial infections and their antibiotic resistance has spurred advancement in the field of material sciences and has guided the development of novel materials with anti-bacterial properties. To address the clinical exigencies, the material of choice should be biodegradable, biocompatible, and able to offer prolonged antibacterial effects. As an attractive option, hydrogels have been explored globally as a potent biomaterial platform that can furnish essential antibacterial attributes owing to its three-dimensional (3D) hydrophilic polymeric network, adequate biocompatibility, and cellular adhesion. The current review focuses on the utilization of different antimicrobial hydrogels based on their sources (natural and synthetic). Further, the review also highlights the strategies for the generation of hydrogels with their advantages and disadvantages and their applications in different biomedical fields. Finally, the prospects in the development of hydrogels-based antimicrobial biomaterials are discussed along with some key challenges encountered during their development and clinical translation.
Collapse
Affiliation(s)
- Anand Singh
- University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Janmay Jai Sharma
- University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Billeswar Mohanta
- University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Ankur Sood
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Anirudh Sharma
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
24
|
Zhuang J, Midgley AC, Wei Y, Liu Q, Kong D, Huang X. Machine-Learning-Assisted Nanozyme Design: Lessons from Materials and Engineered Enzymes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2210848. [PMID: 36701424 DOI: 10.1002/adma.202210848] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/03/2023] [Indexed: 05/11/2023]
Abstract
Nanozymes are nanomaterials that exhibit enzyme-like biomimicry. In combination with intrinsic characteristics of nanomaterials, nanozymes have broad applicability in materials science, chemical engineering, bioengineering, biochemistry, and disease theranostics. Recently, the heterogeneity of published results has highlighted the complexity and diversity of nanozymes in terms of consistency of catalytic capacity. Machine learning (ML) shows promising potential for discovering new materials, yet it remains challenging for the design of new nanozymes based on ML approaches. Alternatively, ML is employed to promote optimization of intelligent design and application of catalytic materials and engineered enzymes. Incorporation of the successful ML algorithms used in the intelligent design of catalytic materials and engineered enzymes can concomitantly facilitate the guided development of next-generation nanozymes with desirable properties. Here, recent progress in ML, its utilization in the design of catalytic materials and enzymes, and how emergent ML applications serve as promising strategies to circumvent challenges associated with time-expensive and laborious testing in nanozyme research and development are summarized. The potential applications of successful examples of ML-aided catalytic materials and engineered enzymes in nanozyme design are also highlighted, with special focus on the unified aims in enhancing design and recapitulation of substrate selectivity and catalytic activity.
Collapse
Affiliation(s)
- Jie Zhuang
- School of Medicine, and State, Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Adam C Midgley
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers, Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Yonghua Wei
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers, Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Qiqi Liu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers, Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Deling Kong
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers, Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers, Science Center for Cell Responses, Nankai University, Tianjin, 300071, China
| |
Collapse
|
25
|
Wang Y, Zhang M, Yan Z, Ji S, Xiao S, Gao J. Metal nanoparticle hybrid hydrogels: the state-of-the-art of combining hard and soft materials to promote wound healing. Theranostics 2024; 14:1534-1560. [PMID: 38389845 PMCID: PMC10879867 DOI: 10.7150/thno.91829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/27/2023] [Indexed: 02/24/2024] Open
Abstract
Wounds represent a grave affliction that profoundly impacts human well-being. Establishing barriers, preventing infections, and providing a conducive microenvironment constitute the crux of wound therapy. Hydrogel, a polymer with an intricate three-dimensional lattice, serves as a potent tool in erecting physical barriers and nurturing an environment conducive to wound healing. This enables effective control over exudation, hemostasis, accelerated wound closure, and diminished scar formation. As a result, hydrogels have gained extensive traction in the realm of wound treatment. Metallic nanoparticle carriers, characterized by their multifaceted responses encompassing acoustics, optics, and electronics, have demonstrated efficacy in wound management. Nevertheless, these carriers encounter challenges associated with swift clearance and nonuniform effectiveness. The hybridization of metallic nanoparticle carriers with hydrogels overcomes the shortcomings inherent in metallic nanoparticle-based wound therapy. This amalgamation not only addresses the limitations but also augments the mechanical robustness of hydrogels. It confers upon them attributes such as environmental responsiveness and multifunctionality, thereby synergizing strengths and compensating for weaknesses. This integration culminates in the precise and intelligent management of wounds. This review encapsulates the structural classifications, design strategies, therapeutic applications, and underlying mechanisms of metal nanoparticle hybrid hydrogels in the context of acute and chronic wound treatment. The discourse delves into the generation of novel or enhanced attributes arising from hybridization and how the current paradigm of wound therapy leverages these attributes. Amidst this continually evolving frontier, the potential of metal nanoparticle hybrid hydrogels to revolutionize wound treatment is underscored.
Collapse
Affiliation(s)
- Yuxiang Wang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Mengya Zhang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Zhenzhen Yan
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Jie Gao
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, People's Republic of China
| |
Collapse
|
26
|
Li J, Yue Z, Tang M, Wang W, Sun Y, Sun T, Chen C. Strategies to Reverse Hypoxic Tumor Microenvironment for Enhanced Sonodynamic Therapy. Adv Healthc Mater 2024; 13:e2302028. [PMID: 37672732 DOI: 10.1002/adhm.202302028] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/26/2023] [Indexed: 09/08/2023]
Abstract
Sonodynamic therapy (SDT) has emerged as a highly effective modality for the treatment of malignant tumors owing to its powerful penetration ability, noninvasiveness, site-confined irradiation, and excellent therapeutic efficacy. However, the traditional SDT, which relies on oxygen availability, often fails to generate a satisfactory level of reactive oxygen species because of the widespread issue of hypoxia in the tumor microenvironment of solid tumors. To address this challenge, various approaches are developed to alleviate hypoxia and improve the efficiency of SDT. These strategies aim to either increase oxygen supply or prevent hypoxia exacerbation, thereby enhancing the effectiveness of SDT. In view of this, the current review provides an overview of these strategies and their underlying principles, focusing on the circulation of oxygen from consumption to external supply. The detailed research examples conducted using these strategies in combination with SDT are also discussed. Additionally, this review highlights the future prospects and challenges of the hypoxia-alleviated SDT, along with the key considerations for future clinical applications. These considerations include the development of efficient oxygen delivery systems, the accurate methods for hypoxia detection, and the exploration of combination therapies to optimize SDT outcomes.
Collapse
Affiliation(s)
- Jialun Li
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Zhengya Yue
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Minglu Tang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Wenxin Wang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Yuan Sun
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin, 150076, P. R. China
| | - Tiedong Sun
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Chunxia Chen
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| |
Collapse
|
27
|
Sun X, Xu X, Yue X, Wang T, Wang Z, Zhang C, Wang J. Nanozymes With Osteochondral Regenerative Effects: An Overview of Mechanisms and Recent Applications. Adv Healthc Mater 2024; 13:e2301924. [PMID: 37633309 DOI: 10.1002/adhm.202301924] [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: 06/17/2023] [Revised: 08/14/2023] [Indexed: 08/28/2023]
Abstract
With the discovery of the intrinsic enzyme-like activity of metal oxides, nanozymes garner significant attention due to their superior characteristics, such as low cost, high stability, multi-enzyme activity, and facile preparation. Notably, in the field of biomedicine, nanozymes primarily focus on disease detection, antibacterial properties, antitumor effects, and treatment of inflammatory conditions. However, the potential for application in regenerative medicine, which primarily addresses wound healing, nerve defect repair, bone regeneration, and cardiovascular disease treatment, is garnering interest as well. This review introduces nanozymes as an innovative strategy within the realm of bone regenerative medicine. The primary focus of this approach lies in the facilitation of osteochondral regeneration through the modulation of the pathological microenvironment. The catalytic mechanisms of four types of representative nanozymes are first discussed. The pathological microenvironment inhibiting osteochondral regeneration, followed by summarizing the therapy mechanism of nanozymes to osteochondral regeneration barriers is introduced. Further, the therapeutic potential of nanozymes for bone diseases is included. To improve the therapeutic efficiency of nanozymes and facilitate their clinical translation, future potential applications in osteochondral diseases are also discussed and some significant challenges addressed.
Collapse
Affiliation(s)
- Xueheng Sun
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, 200438, China
| | - Xiang Xu
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| | - Xiaokun Yue
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| | - Tianchang Wang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| | - Zhaofei Wang
- Department of Orthopaedic Surgery, Shanghai ZhongYe Hospital, Genertec Universal Medical Group, Shanghai, 200941, China
| | - Changru Zhang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
- Institute of Translational Medicine, Shanghai Jiaotong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Jinwu Wang
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, 200438, China
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Rd, Shanghai, 200011, China
| |
Collapse
|
28
|
Sun T, Xiao S, Wang M, Xie Q, Zhang L, Gong M, Zhang D, Zhou C. Reactive Oxygen Species Scavenging Nanozymes: Emerging Therapeutics for Acute Liver Injury Alleviation. Int J Nanomedicine 2023; 18:7901-7922. [PMID: 38148856 PMCID: PMC10750792 DOI: 10.2147/ijn.s435544] [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: 09/13/2023] [Accepted: 12/05/2023] [Indexed: 12/28/2023] Open
Abstract
Acute liver injury (AIL), a fatal clinical disease featured with a swift deterioration of hepatocyte functions in the short term, has emerged as a serious public health issues that warrants attention. However, the effectiveness of existing small molecular antioxidants and anti-inflammatory medications in alleviating AIL remains uncertain. The unique inherent structural characteristics of liver confer it a natural propensity for nanoparticle capture, which present an opportunity to exploit in the formulation of nanoscale therapeutic agents, enabling their selective accumulation in the liver and thereby facilitating targeted therapeutic interventions. Significantly increased reactive oxygen species (ROS) accumulation and inflammation response have been evidenced to play crucial roles in occurrence and development of AIL. Nanozymes with ROS-scavenging capacities have demonstrated considerable promise in ROS elimination and inflammation regulation, thereby offering an appealing therapeutic instrument for the management of acute liver injury. In this review, the mechanisms of different type of ALI were summarized. In addition, we provide a comprehensive summary and review of the available ROS-scavenging nanozymes, including transition metal-based nanozymes, noble metal nanozymes, carbon-based nanozymes, and some other nanozymes. Furthermore, the challenges still need to be solved in the field of ROS-scavenging nanozymes for ALI alleviation are also discussed.
Collapse
Affiliation(s)
- Tao Sun
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, People’s Republic of China
| | - Shilin Xiao
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Miaomiao Wang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Qian Xie
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Liang Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Mingfu Gong
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Dong Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Chunyu Zhou
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
29
|
Zhang C, Zhou X, Wang D, Hao L, Zeng Z, Su L. Hydrogel-Loaded Exosomes: A Promising Therapeutic Strategy for Musculoskeletal Disorders. J Clin Pharm Ther 2023; 2023:1-36. [DOI: 10.1155/2023/1105664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
Clinical treatment strategies for musculoskeletal disorders have been a hot research topic. Accumulating evidence suggests that hydrogels loaded with MSC-derived EVs show great potential in improving musculoskeletal injuries. The ideal hydrogels should be capable of promoting the development of new tissues and simulating the characteristics of target tissues, with the properties matching the cell-matrix constituents of autologous tissues. Although there have been numerous reports of hydrogels loaded with MSC-derived EVs for the repair of musculoskeletal injuries, such as intervertebral disc injury, tendinopathy, bone fractures, and cartilage injuries, there are still many hurdles to overcome before the clinical application of modified hydrogels. In this review, we focus on the advantages of the isolation technique of EVs in combination with different types of hydrogels. In this context, the efficacy of hydrogels loaded with MSC-derived EVs in different musculoskeletal injuries is discussed in detail to provide a reference for the future application of hydrogels loaded with MSC-derived EVs in the clinical treatment of musculoskeletal injuries.
Collapse
Affiliation(s)
- Chunyu Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Dongxue Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Li Hao
- Shougang Technician College, Nursing School, Beijing 100043, China
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| | - Zhipeng Zeng
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
- Shougang Technician College, Nursing School, Beijing 100043, China
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| | - Lei Su
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| |
Collapse
|
30
|
Zhu H, Liu F, Zhai X, Tong Z, Li H, Dong W, Wei W, Teng C. Revisiting matrix hydrogel composed of gelatin and hyaluronic acid and its application in cartilage regeneration. Biochem Biophys Res Commun 2023; 681:97-105. [PMID: 37774575 DOI: 10.1016/j.bbrc.2023.09.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/07/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
With the increasing incidence of knee osteoarthritis (KOA), the reparation of cartilage defects is gaining more attention. Given that tissue integration plays a critical role in repairing cartilage defects, tissue adhesive hydrogels are highly needed in clinics. We constructed a biomacromolecule-based bioadhesive matrix hydrogel and applied it to promote cartilage regeneration. The hydrogel was composed of methacrylate gelatin and N-(2-aminoethyl)-4-(4-(hydroxymethyl)-2-methoxy-5-nitroso) butyl amide modified hyaluronic acid (HANB). The methacrylate gelatin provided a stable hydrogel network as a scaffold, and the HANB served as a tissue-adhesive agent and could be favorable for the chondrogenesis of stem cells. Additionally, the chemically modified HA increased the swelling ratio and compressive modulus of the hydrogels. The results of our in vitro study revealed that the hydrogel was compatible with bone marrow stromal cells. In vivo, the hyaluronic-acid-containing hydrogels were found to promote articular cartilage regeneration in the defect site. Therefore, this biomaterial provides promising potential for cartilage repair.
Collapse
Affiliation(s)
- Huangrong Zhu
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Fengling Liu
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China; School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, 210094, China
| | - Xinrang Zhai
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China; School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, 210094, China
| | - Zhicheng Tong
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Huimin Li
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Wei Dong
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, 210094, China
| | - Wei Wei
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, China.
| | - Chong Teng
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China.
| |
Collapse
|
31
|
Jin Y, Zhang J, Xu Y, Yi K, Li F, Zhou H, Wang H, Chan HF, Lao YH, Lv S, Tao Y, Li M. Stem cell-derived hepatocyte therapy using versatile biomimetic nanozyme incorporated nanofiber-reinforced decellularized extracellular matrix hydrogels for the treatment of acute liver failure. Bioact Mater 2023; 28:112-131. [PMID: 37250866 PMCID: PMC10209199 DOI: 10.1016/j.bioactmat.2023.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 04/07/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Reactive oxygen species (ROS)-associated oxidative stress, inflammation storm, and massive hepatocyte necrosis are the typical manifestations of acute liver failure (ALF), therefore specific therapeutic interventions are essential for the devastating disease. Here, we developed a platform consisting of versatile biomimetic copper oxide nanozymes (Cu NZs)-loaded PLGA nanofibers (Cu NZs@PLGA nanofibers) and decellularized extracellular matrix (dECM) hydrogels for delivery of human adipose-derived mesenchymal stem/stromal cells-derived hepatocyte-like cells (hADMSCs-derived HLCs) (HLCs/Cu NZs@fiber/dECM). Cu NZs@PLGA nanofibers could conspicuously scavenge excessive ROS at the early stage of ALF, and reduce the massive accumulation of pro-inflammatory cytokines, herein efficiently preventing the deterioration of hepatocytes necrosis. Moreover, Cu NZs@PLGA nanofibers also exhibited a cytoprotection effect on the transplanted HLCs. Meanwhile, HLCs with hepatic-specific biofunctions and anti-inflammatory activity acted as a promising alternative cell source for ALF therapy. The dECM hydrogels further provided the desirable 3D environment and favorably improved the hepatic functions of HLCs. In addition, the pro-angiogenesis activity of Cu NZs@PLGA nanofibers also facilitated the integration of the whole implant with the host liver. Hence, HLCs/Cu NZs@fiber/dECM performed excellent synergistic therapeutic efficacy on ALF mice. This strategy using Cu NZs@PLGA nanofiber-reinforced dECM hydrogels for HLCs in situ delivery is a promising approach for ALF therapy and shows great potential for clinical translation.
Collapse
Affiliation(s)
- Yuanyuan Jin
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Jiabin Zhang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Fenfang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Huicong Zhou
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| |
Collapse
|
32
|
Chen G, Wang Q, Zhu Y, Zhao M, Ma S, Bai Y, Wang J, Zou M, Cheng G. Molecularly engineered dual-network photothermal hydrogel delivery system with enhanced mechanical properties, antibacterial ability and angiogenic effect for accelerating wound healing. J Mech Behav Biomed Mater 2023; 146:106081. [PMID: 37651758 DOI: 10.1016/j.jmbbm.2023.106081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/14/2023] [Accepted: 08/20/2023] [Indexed: 09/02/2023]
Abstract
Bacterial infection caused by trauma and chronic wounds in the most mobile area remains a challenge in clinic. It is difficult to achieve the synergistic effects of antibacterial capacity and skin regeneration using conventional therapeutic methods. Developing a multi-functional hydrogel dressing that can cope with the complex wound environment will contribute to the healing and therapeutic effects. In this work, a novel Cur@PAM/TA-Cu photothermal hydrogel delivery system was prepared by engineering tannic acid (TA) into covalent cross-linked polyacrylamide (PAM) on which the chelating tannic acid-copper metal-polyphenolic network (TA-Cu MPN) was imposed to form dual-crosslinked networks, and the natural medicine curcumin was loaded eventually. The molecularly engineered dual-crosslinked networks resulted in enhanced mechanical properties including bio-adhesion, tensile strength and self-healing, which made the hydrogel suitable for dynamic wound and various application scenarios. In addition, the excellent photothermal capacity, antioxidant effect and biocompatibility of the hydrogel were demonstrated. Notably, this curcumin loaded photothermal hydrogel exhibited superior antibacterial capacity (almost 100% killing ratio to E. coli and S. aureus) under 808 nm laser irradiation. Meanwhile, the in vivo wound healing experiment results revealed that the anti-inflammation and proangiogenic effect of Cur@PAM/TA-Cu hydrogel successfully shortened the healing time of wound and the reconstruction of skin structure and function. Thus, this dual-crosslinked multi-functional hydrogel delivery system is a promising wound dressing for accelerating wound healing.
Collapse
Affiliation(s)
- Guo Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Qiaoqiao Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Yumeng Zhu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Minqian Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Siyuan Ma
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Yifeng Bai
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Jingfeng Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Meijuan Zou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Gang Cheng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China.
| |
Collapse
|
33
|
Lyu Y, Liu Y, He H, Wang H. Application of Silk-Fibroin-Based Hydrogels in Tissue Engineering. Gels 2023; 9:gels9050431. [PMID: 37233022 DOI: 10.3390/gels9050431] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023] Open
Abstract
Silk fibroin (SF) is an excellent protein-based biomaterial produced by the degumming and purification of silk from cocoons of the Bombyx mori through alkali or enzymatic treatments. SF exhibits excellent biological properties, such as mechanical properties, biocompatibility, biodegradability, bioabsorbability, low immunogenicity, and tunability, making it a versatile material widely applied in biological fields, particularly in tissue engineering. In tissue engineering, SF is often fabricated into hydrogel form, with the advantages of added materials. SF hydrogels have mostly been studied for their use in tissue regeneration by enhancing cell activity at the tissue defect site or counteracting tissue-damage-related factors. This review focuses on SF hydrogels, firstly summarizing the fabrication and properties of SF and SF hydrogels and then detailing the regenerative effects of SF hydrogels as scaffolds in cartilage, bone, skin, cornea, teeth, and eardrum in recent years.
Collapse
Affiliation(s)
- Yihan Lyu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yusheng Liu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Houzhe He
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hongmei Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
34
|
Zhang P, Chen J, Sun Y, Cao Z, Zhang Y, Mo Q, Yao Q, Zhang W. A 3D multifunctional bi-layer scaffold to regulate stem cell behaviors and promote osteochondral regeneration. J Mater Chem B 2023; 11:1240-1261. [PMID: 36648128 DOI: 10.1039/d2tb02203f] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Osteochondral defect (OCD) regeneration remains a great challenge. Recently, multilayer scaffold simulating native osteochondral structures have aroused broad interest in osteochondral tissue engineering. Here, we developed a 3D multifunctional bi-layer scaffold composed of a kartogenin (KGN)-loaded GelMA hydrogel (GelMA/KGN) as an upper layer mimicking a cartilage-specific extracellular matrix and a hydroxyapatite (HA)-coated 3D printed polycaprolactone porous scaffold (PCL/HA) as a lower layer simulating subchondral bone. The bi-layer scaffolds were subsequently modified with tannic acid (TA) prime-coating and E7 peptide conjugation (PCL/HA-GelMA/KGN@TA/E7) to regulate endogenous stem cell behaviors and exert antioxidant activity for enhanced osteochondral regeneration. In vitro, the scaffolds could support cell attachment and proliferation, and enhance the chondrogenic and osteogenic differentiation capacity of bone marrow-derived mesenchymal stem cells (BMSCs) in a specific layer. Besides, the incorporation of TA/E7 significantly increased the biological activity of the bi-layer scaffolds including the pro-migratory effect, antioxidant activity, and the maintenance of cell viability against oxidative stress. In vivo, the developed bi-layer scaffolds enhanced the simultaneous regeneration of cartilage and subchondral bone when implanted into a rabbit OCD model through macroscopic, micro-CT, and histological evaluation. Taken together, these investigations demonstrated that the 3D multifunctional bi-layer scaffolds could provide a suitable microenvironment for endogenous stem cells, and promote in situ osteochondral regeneration, showing great potential for the clinical treatment of OCD.
Collapse
Affiliation(s)
- Po Zhang
- Department of Orthopedic Surgery, Digital Medicine Institute, Nanjing Medical University Nanjing Hospital, No. 68 Changle Road, Nanjing 210006, P. R. China.
- School of Medicine, Southeast University, 210009, Nanjing, China.
| | - Jialin Chen
- School of Medicine, Southeast University, 210009, Nanjing, China.
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Yuzhi Sun
- Department of Orthopedic Surgery, Digital Medicine Institute, Nanjing Medical University Nanjing Hospital, No. 68 Changle Road, Nanjing 210006, P. R. China.
- School of Medicine, Southeast University, 210009, Nanjing, China.
| | - Zhicheng Cao
- Department of Orthopedic Surgery, Digital Medicine Institute, Nanjing Medical University Nanjing Hospital, No. 68 Changle Road, Nanjing 210006, P. R. China.
- School of Medicine, Southeast University, 210009, Nanjing, China.
| | - Yanan Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China.
| | - Qingyun Mo
- School of Medicine, Southeast University, 210009, Nanjing, China.
| | - Qingqiang Yao
- Department of Orthopedic Surgery, Digital Medicine Institute, Nanjing Medical University Nanjing Hospital, No. 68 Changle Road, Nanjing 210006, P. R. China.
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China.
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| |
Collapse
|
35
|
Luo Q, Shao N, Zhang AC, Chen CF, Wang D, Luo LP, Xiao ZY. Smart Biomimetic Nanozymes for Precise Molecular Imaging: Application and Challenges. Pharmaceuticals (Basel) 2023; 16:249. [PMID: 37259396 PMCID: PMC9965384 DOI: 10.3390/ph16020249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 04/06/2024] Open
Abstract
New nanotechnologies for imaging molecules are widely being applied to visualize the expression of specific molecules (e.g., ions, biomarkers) for disease diagnosis. Among various nanoplatforms, nanozymes, which exhibit enzyme-like catalytic activities in vivo, have gained tremendously increasing attention in molecular imaging due to their unique properties such as diverse enzyme-mimicking activities, excellent biocompatibility, ease of surface tenability, and low cost. In addition, by integrating different nanoparticles with superparamagnetic, photoacoustic, fluorescence, and photothermal properties, the nanoenzymes are able to increase the imaging sensitivity and accuracy for better understanding the complexity and the biological process of disease. Moreover, these functions encourage the utilization of nanozymes as therapeutic agents to assist in treatment. In this review, we focus on the applications of nanozymes in molecular imaging and discuss the use of peroxidase (POD), oxidase (OXD), catalase (CAT), and superoxide dismutase (SOD) with different imaging modalities. Further, the applications of nanozymes for cancer treatment, bacterial infection, and inflammation image-guided therapy are discussed. Overall, this review aims to provide a complete reference for research in the interdisciplinary fields of nanotechnology and molecular imaging to promote the advancement and clinical translation of novel biomimetic nanozymes.
Collapse
Affiliation(s)
| | | | | | | | | | - Liang-Ping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Ze-Yu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| |
Collapse
|
36
|
Li D, Dai D, Xiong G, Lan S, Zhang C. Metal-Based Nanozymes with Multienzyme-Like Activities as Therapeutic Candidates: Applications, Mechanisms, and Optimization Strategy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205870. [PMID: 36513384 DOI: 10.1002/smll.202205870] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/21/2022] [Indexed: 06/17/2023]
Abstract
Most nanozymes in development for medical applications only exhibit single-enzyme-like activity, and are thus limited by insufficient catalytic activity and dysfunctionality in complex pathological microenvironments. To overcome the impediments of limited substrate availabilities and concentrations, some metal-based nanozymes may mimic two or more activities of natural enzymes to catalyze cascade reactions or to catalyze multiple substrates simultaneously, thereby amplifying catalysis. Metal-based nanozymes with multienzyme-like activities (MNMs) may adapt to dissimilar catalytic conditions to exert different enzyme-like effects. These multienzyme-like activities can synergize to realize "self-provision of the substrate," in which upstream catalysts produce substrates for downstream catalytic reactions to overcome the limitation of insufficient substrates in the microenvironment. Consequently, MNMs exert more potent antitumor, antibacterial, and anti-inflammatory effects in preclinical models. This review summarizes the cellular effects and underlying mechanisms of MNMs. Their potential medical utility and optimization strategy from the perspective of clinical requirements are also discussed, with the aim to provide a theoretical reference for the design, development, and therapeutic application of their catalytic effects.
Collapse
Affiliation(s)
- Dan Li
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Danni Dai
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Gege Xiong
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Shuquan Lan
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Chao Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
37
|
Cai J, Liu LF, Qin Z, Liu S, Wang Y, Chen Z, Yao Y, Zheng L, Zhao J, Gao M. Natural Morin-Based Metal Organic Framework Nanoenzymes Modulate Articular Cavity Microenvironment to Alleviate Osteoarthritis. RESEARCH (WASHINGTON, D.C.) 2023; 6:0068. [PMID: 36930778 PMCID: PMC10013961 DOI: 10.34133/research.0068] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
Osteoarthritis (OA) is always characterized as excessive reactive oxygen species (ROS) inside articular cavity. Mimicking natural metalloenzymes with metal ions as the active centers, stable metal organic framework (MOF) formed by natural polyphenols and metal ions shows great potential in alleviating inflammatory diseases. Herein, a series of novel copper-morin-based MOF (CuMHs) with different molar ratios of Cu2+ and MH were employed to serve as ROS scavengers for OA therapy. As a result, CuMHs exhibited enhanced dispersion in aqueous solution, improved biocompatibility, and efficient ROS-scavenging ability compared to MH. On the basis of H2O2-stimulated chondrocytes, intracellular ROS levels were efficiently declined and cell death was prevented after treated by Cu6MH (Cu2+ and MH molar ratio of 6:1). Meanwhile, Cu6MH also exhibited efficient antioxidant and anti-inflammation function by down-regulating the expression of IL6, MMP13, and MMP3, and up-regulating cartilage specific gene expression as well. Importantly, Cu6MH could repair mitochondrial function by increasing mitochondrial membrane potential, reducing the accumulation of calcium ions, as well as promoting ATP content production. In OA joint model, intra-articular (IA) injected Cu6MH suppressed the progression of OA. It endowed that Cu6MH might be promising nanoenzymes for the prevention and treatment of various inflammatory diseases.
Collapse
Affiliation(s)
- Jinhong Cai
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Lian-Feng Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Department of Ultrasound, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Zainen Qin
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Shuhan Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Yonglin Wang
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Department of Orthopedics, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Zhengrong Chen
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Yi Yao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Li Zheng
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Jinmin Zhao
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Department of Orthopedics, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Ming Gao
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
38
|
Hu H, Huang X, Dai Y, Zhu K, Ye X, Meng S, Zhang Q, Xie X. Organic metal matrix Mil-88a nano-enzyme for joint repair in the osteoarthritis mouse model. Front Bioeng Biotechnol 2023; 11:1164942. [PMID: 37187885 PMCID: PMC10175628 DOI: 10.3389/fbioe.2023.1164942] [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: 02/13/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Introduction: In this paper we tried to conduct a novel nanomaterial strategy to overcome osteoarthritis (OA) in a mouse model. Methods: In this regard, after synthesizing the Mil-88a nanozyme, as a certain Fe-MOF, its toxic effects were detected by CCK-8 method and live-dead staining. The OA model of mouse was constructed, and paraffin sections of joints were taken for histological evaluation. In addition, immunofluorescence and immunohistochemistry were used to identify the OA progression and OARSI was used to evaluate the OA grades. We observed that Mil-88a could be easily synthesized and has high biocompatibility. Results: We observed that Mil-88a could significantly promote the expression of OA anabolism-related genes such as Col2 and also significantly inhibit the expression of OA catabolism-related genes MMP13. Besides, we observed better OARSI score in animals treated with Mil-88a nano-enzyme loading on organic metal matrix. Discussion: Overall, Mil-88a nano-enzyme could be used as a novel strategy to treat OA.
Collapse
|
39
|
An Injectable Hydrogel Scaffold Loaded with Dual-Drug/Sustained-Release PLGA Microspheres for the Regulation of Macrophage Polarization in the Treatment of Intervertebral Disc Degeneration. Int J Mol Sci 2022; 24:ijms24010390. [PMID: 36613833 PMCID: PMC9820357 DOI: 10.3390/ijms24010390] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/10/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Due to the unique physical characteristics of intervertebral disc degeneration (IVDD) and the pathological microenvironment that it creates, including inflammation and oxidative stress, effective self-repair is impossible. During the process of intervertebral disc degeneration, there is an increase in the infiltration of M1 macrophages and the secretion of proinflammatory cytokines. Here, we designed a novel injectable composite hydrogel scaffold: an oligo [poly (ethylene glycol) fumarate]/sodium methacrylate (OPF/SMA) hydrogel scaffold loaded with dual-drug/sustained-release PLGA microspheres containing IL-4 (IL-4-PLGA) and kartogenin (KGN-PLGA). This scaffold exhibited good mechanical properties and low immunogenicity while also promoting the sustained release of drugs. By virtue of the PLGA microspheres loaded with IL-4 (IL-4-PLGA), the composite hydrogel scaffold induced macrophages to transition from the M1 phenotype into the M2 phenotype during the early induced phase and simultaneously exhibited a continuous anti-inflammatory effect through the PLGA microspheres loaded with kartogenin (KGN-PLGA). Furthermore, we investigated the mechanisms underlying the immunomodulatory and anti-inflammatory effects of the composite hydrogel scaffold. We found that the scaffold promoted cell proliferation and improved cell viability in vitro. While ensuring mechanical strength, this composite hydrogel scaffold regulated the local inflammatory microenvironment and continuously repaired tissue in the nucleus pulposus via the sequential release of drugs in vivo. When degenerative intervertebral discs in a rat model were injected with the scaffold, there was an increase in the proportion of M2 macrophages in the inflammatory environment and higher expression levels of type II collagen and aggrecan; this was accompanied by reduced levels of MMP13 expression, thus exhibiting long-term anti-inflammatory effects. Our research provides a new strategy for promoting intervertebral disc tissue regeneration and a range of other inflammatory diseases.
Collapse
|
40
|
Wang Q, Luo Z, Wu YL, Li Z. Recent Advances in Enzyme‐Based Biomaterials Toward Diabetic Wound Healing. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Qi Wang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology School of Pharmaceutical Sciences Xiamen University Xiamen 361102 China
| | - Zheng Luo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology School of Pharmaceutical Sciences Xiamen University Xiamen 361102 China
- Institute of Materials Research and Engineering A*STAR (Agency for Science, Technology and Research) 2 Fusionopolis Way Innovis, #08-03 Singapore 138634 Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology School of Pharmaceutical Sciences Xiamen University Xiamen 361102 China
| | - Zibiao Li
- Institute of Materials Research and Engineering A*STAR (Agency for Science, Technology and Research) 2 Fusionopolis Way Innovis, #08-03 Singapore 138634 Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2) Agency for Science, Technology and Research (A*STAR) 2 Fusionopolis Way Singapore 138634 Singapore
- Department of Materials Science and Engineering National University of Singapore 9 Engineering Drive 1 Singapore 117576 Singapore
| |
Collapse
|
41
|
Jin P, Liu L, Chen X, Cheng L, Zhang W, Zhong G. Applications and prospects of different functional hydrogels in meniscus repair. Front Bioeng Biotechnol 2022; 10:1082499. [PMID: 36568293 PMCID: PMC9773848 DOI: 10.3389/fbioe.2022.1082499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
The meniscus is a kind of fibrous cartilage structure that serves as a cushion in the knee joint to alleviate the mechanical load. It is commonly injured, but it cannot heal spontaneously. Traditional meniscectomy is not currently recommended as this treatment tends to cause osteoarthritis. Due to their good biocompatibility and versatile regulation, hydrogels are emerging biomaterials in tissue engineering. Hydrogels are excellent candidates in meniscus rehabilitation and regeneration because they are fine-tunable, easily modified, and capable of delivering exogenous drugs, cells, proteins, and cytokines. Various hydrogels have been reported to work well in meniscus-damaged animals, but few hydrogels are effective in the clinic, indicating that hydrogels possess many overlooked problems. In this review, we summarize the applications and problems of hydrogels in extrinsic substance delivery, meniscus rehabilitation, and meniscus regeneration. This study will provide theoretical guidance for new therapeutic strategies for meniscus repair.
Collapse
Affiliation(s)
- Pan Jin
- Health Science Center, Yangtze University, Jingzhou, China,Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, China,*Correspondence: Pan Jin, ; Gang Zhong,
| | - Lei Liu
- Articular Surgery, The Second Nanning People’s Hospital (Third Affiliated Hospital of Guangxi Medical University), Nanning, China
| | - Xichi Chen
- Health Science Center, Yangtze University, Jingzhou, China
| | - Lin Cheng
- Health Science Center, Yangtze University, Jingzhou, China
| | - Weining Zhang
- Health Science Center, Yangtze University, Jingzhou, China
| | - Gang Zhong
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China,*Correspondence: Pan Jin, ; Gang Zhong,
| |
Collapse
|