1
|
Choi S, Choi H, Chung JW, Kim SH. Injectable Endoplasmin-Loaded Lipid Nanoparticles-Hydrogel Composite for Cartilage Regeneration. Tissue Eng Regen Med 2025; 22:409-424. [PMID: 39992620 PMCID: PMC12122951 DOI: 10.1007/s13770-024-00698-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/12/2024] [Accepted: 12/22/2024] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Endoplasmin (ENPL), a heat shock protein 90 family member, promotes chondrogenic differentiation of stem cells by inhibiting ERK1/2 phosphorylation and inducing endoplasmic reticulum stress. However, its large size limits cellular uptake and therapeutic potential. To overcome this challenge, a cationic lipid nanoparticle (C_LNP) system was designed to deliver ENPL intracellularly, enhancing its effects on human tonsil-derived mesenchymal stem cells (hTMSCs). METHODS ENPL-loaded cationic lipid nanoparticles (ENPL_C_LNP) were synthesized to facilitate intracellular ENPL delivery. The delivery efficiency and cytotoxicity were assessed in vitro using hTMSCs. Additionally, ENPL_C_LNPs were incorporated into a hyaluronic acid and chondroitin sulfate-based injectable hydrogel and tested for chondrogenic differentiation potential in a mouse subcutaneous model. RESULTS ENPL_C_LNP achieved over 80% intracellular protein delivery efficiency with no cytotoxic effects. Co-cultured hTMSCs exhibited increased glycosaminoglycans (GAGs) and collagen expression over 21 days. In vivo, the hydrogel-embedded ENPL_C_LNP system enabled stable cartilage differentiation, evidenced by abundant cartilage-specific lacuna structures in regenerated tissue. CONCLUSION Combining ENPL_C_LNP with an injectable hydrogel scaffold supports chondrogenic differentiation and cartilage regeneration, offering a promising strategy for cartilage tissue engineering.
Collapse
Affiliation(s)
- Sumi Choi
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan, 49315, Republic of Korea
| | - Hyeongrok Choi
- Department of Biomedical Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Jin Woong Chung
- Department of Biomedical Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
2
|
Liu Y, Chen ZJ, Fei Y, Yu X, Chen G. Investigating the therapeutic potential of epigallocatechin gallate (EGCG) for chronic pain management: mechanisms, applications, and future perspectives. Fitoterapia 2025; 184:106646. [PMID: 40446933 DOI: 10.1016/j.fitote.2025.106646] [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: 02/08/2025] [Revised: 05/12/2025] [Accepted: 05/27/2025] [Indexed: 06/02/2025]
Abstract
Chronic pain, a widespread condition impacting a large segment of the global population, involves persistent and diverse pain types including nociceptive, neuropathic, and the more recently defined nociplastic pain. This review explores the potential of Epigallocatechin gallate (EGCG), a key bioactive compound in green tea, known for its extensive biological activities that aid in pain management. Highlighting its anti-inflammatory, antioxidant, and neuroprotective effects, EGCG is posited as a promising alternative to conventional pain medications, which are often associated with significant side effects and dependency risks. Despite its therapeutic promise, EGCG's clinical application is hampered by poor bioavailability and stability. By outlining the current state of research and identifying gaps for future investigation, this review underscores the importance of integrating EGCG into a broader pain management paradigm. It advocates for extensive clinical trials to confirm EGCG's efficacy and safety in humans, aiming to establish it as a part of an integrative strategy for treating chronic pain, thus potentially reducing the reliance on conventional pharmacotherapy.
Collapse
Affiliation(s)
- Yue Liu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China
| | - Zheng-Jie Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China
| | - Yue Fei
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China
| | - Xin Yu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China; Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China.
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China.
| |
Collapse
|
3
|
Xu X, Huang X, Lin S, Yu G, Xiao J, Wang J, Song C, Khalilov F, Issaro N, Xu N, Gao S. Hyaluronic acid/ε-polylysine hydrogel enriched with Saussurea involucrata polysaccharide for improved skin dryness induced by ultraviolet radiation. Int J Biol Macromol 2025; 308:142718. [PMID: 40174445 DOI: 10.1016/j.ijbiomac.2025.142718] [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: 01/04/2025] [Revised: 03/21/2025] [Accepted: 03/30/2025] [Indexed: 04/04/2025]
Abstract
Ultraviolet B (UVB) radiation exposure is a major contributor to skin photoaging, ultimately resulting in dryness. Conventional treatments for this condition frequently exhibit limited efficacy due to poor skin penetration and are associated with significant side effects. Saussurea involucrate polysaccharide (SIP) has been shown to alleviate photoaging-induced dry skin; however, its effectiveness is limited due to low drug utilization and the need for frequent dosing. In this study, we developed hyaluronic acid (HA) and ε-polylysine (ε-PLL) hydrogels designed to enhance skin hydration and enable sustained drug release. The optimal formulation, designated A5E3, contained 5 % HA plus 3 % ε-PLL. The sustained release of SIP from A5E3 demonstrated superior therapeutic effects in a mouse model of skin photoaging, including reduced inflammation and apoptosis, as well as enhanced keratinocyte differentiation and lipid production. The A5E3 hydrogel not only enhanced the drug utilization of SIP but also provided a sustained moisturizing environment, promoting faster skin repair. These findings support the development of skincare products with superior moisturizing and reparative properties, underscoring the potential of hydrogels as effective carriers for treating UVB-induced skin dryness and damage.
Collapse
Affiliation(s)
- Xiashun Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Xulong Huang
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Shisheng Lin
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Guangdong Yu
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Jiali Xiao
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Junchao Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chengyang Song
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Farid Khalilov
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Nipatha Issaro
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Nuo Xu
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China.
| | - Shuang Gao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
4
|
Liang W, Yang R, Qin L, Liang T, Chen W. Current Status and Perspectives of Research on Polymer Hydrogels in the Treatment and Protection of Osteoarthritis. Macromol Biosci 2025:e2500016. [PMID: 40271818 DOI: 10.1002/mabi.202500016] [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: 01/07/2025] [Revised: 04/04/2025] [Indexed: 04/25/2025]
Abstract
Arthritis is a degenerative disease characterized by chronic cartilage degeneration. It affects hundreds of millions of people worldwide and often has serious consequences such as joint pain and swelling, limited mobility, and joint deformity. However, conventional treatments still struggle to achieve satisfactory results. Finding more effective treatments for arthritis remains an important clinical challenge. As hydrogels have a unique 3D spatial mesh structure, significant material interaction ability, adjustable mechanical properties, and good biodegradability, they can provide a suitable cellular or tissue microenvironment, and their potential in scaffolding effect, lubrication, anti-inflammatory effect, or drug or cellular delivery is expected to be a potent therapeutic approach for the treatment of osteoarthritis. In this review, three aspects of hydrogel products for osteoarthritis treatment are comprehensively summarized and discussed, namely, material selection and gel design, exploration of cross-linking mechanisms, and mechanisms of hydrogel therapy for osteoarthritis, and focus on the advantages and limitations of their clinical applications, which point out the direction of the development strategy of innovative products in this field, applied research, and clinical transformation.
Collapse
Affiliation(s)
- Wanjun Liang
- School of Pharmaceutical Sciences, Institute of Materia Medica, Shandong Academy of Medical Sciences, State Key Laboratory of Advanced Drug Delivery and Release System, Shandong First Medical University, Jinan, 250117, China
| | - Rui Yang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Lijing Qin
- School of Pharmaceutical Sciences, Institute of Materia Medica, Shandong Academy of Medical Sciences, State Key Laboratory of Advanced Drug Delivery and Release System, Shandong First Medical University, Jinan, 250117, China
| | - Tongjuan Liang
- School of Pharmaceutical Sciences, Institute of Materia Medica, Shandong Academy of Medical Sciences, State Key Laboratory of Advanced Drug Delivery and Release System, Shandong First Medical University, Jinan, 250117, China
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
5
|
Jin X, Yue Y, Hu H, Lv S. Traditional Chinese Medicine-Loaded Hydrogels: An Emerging Strategy for the Treatment of Bone Infections. Pharmaceutics 2025; 17:514. [PMID: 40284509 PMCID: PMC12030446 DOI: 10.3390/pharmaceutics17040514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Bone infection is a disease that seriously affects patients' quality of life and physical health. Traditional treatment methods have many drawbacks. Hydrogels loaded with Traditional Chinese Medicine (TCM), as an emerging treatment strategy, combine the advantages of good biocompatibility of hydrogels, adjustable drug release performance, and multi-target synergistic treatment of TCM, showing great application potential. This article elaborates in detail on the research progress of hydrogels loaded with TCM for the treatment of bone infections, including the classification and characteristics of hydrogels, the mechanism of action of TCM in the treatment of bone infections, the preparation methods of hydrogels loaded with TCM, application examples, advantages, and the challenges and prospects faced. The aim is to provide new ideas and references for the clinical treatment of bone infections.
Collapse
Affiliation(s)
| | | | - Huaanzi Hu
- School of Pharmacy, Changzhou University, Changzhou 213164, China; (X.J.); (Y.Y.)
| | - Songwei Lv
- School of Pharmacy, Changzhou University, Changzhou 213164, China; (X.J.); (Y.Y.)
| |
Collapse
|
6
|
Petit N, Chang YYJ, Lobianco FA, Hodgkinson T, Browne S. Hyaluronic acid as a versatile building block for the development of biofunctional hydrogels: In vitro models and preclinical innovations. Mater Today Bio 2025; 31:101596. [PMID: 40083836 PMCID: PMC11903855 DOI: 10.1016/j.mtbio.2025.101596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
Hyaluronic acid (HyA) is a non-sulphated linear polysaccharide found abundantly in the extracellular matrix, known for its biocompatibility and versatility in tissue engineering. Chemical modifications of HyA, including methacrylate, acrylate, click chemistry, norbornene, or host-guest chemistry, are necessary for the formation of stable hydrogels with tuneable biophysical characteristics. These modifications enable precise control over stiffness, swelling, degradation, and advanced functionalities such as shear-thinning, self-healing, and injectability. Functionalisation further enhances hydrogel bioactivity, enabling controlled cell adhesion, modulation of cell behaviour, hydrogel degradation, and release profiles, as well as inflammation modulation or bacterial growth inhibition. These are achieved by conjugating proteins, peptides, antibodies, or reactive chemical groups. HyA hydrogels find broad applications both in vitro and in vivo. In vitro, HyA-based hydrogels can support the development of models to understand fundamental processes in health and mechanisms behind disease progression, serving as highly tuneable extracellular matrix mimetics. As therapeutic interventions, injectable or implantable HyA-based hydrogels have been developed to repair a range of tissues, including cartilage, bone, muscle, and skin defects. However, issues remain to be addressed before widespread adoption of HyA-based hydrogels as clinical options. Future innovations for HyA hydrogels include its establishment as an enabling technology for the delivery of novel therapeutics, with a particular focus on immunomodulatory molecules, and the development of more dynamic, tissue-mimetic HyA-based hydrogels.
Collapse
Affiliation(s)
- Noémie Petit
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Yu-yin Joanne Chang
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Franz Acker Lobianco
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
| | - Tom Hodgkinson
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
| | - Shane Browne
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
7
|
Wu X, Tao W, Lan Z, Tian Y, Zhong Z, Wang J, Li J, Liu X, Zhang X, Wang Y, Wang J, Zhang B, Du Y, Zhang S. pH-Responsive Engineered Exosomes Enhance Endogenous Hyaluronan Production by Reprogramming Chondrocytes for Cartilage Repair. Adv Healthc Mater 2025; 14:e2405126. [PMID: 40042438 DOI: 10.1002/adhm.202405126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/20/2025] [Indexed: 04/18/2025]
Abstract
Trauma or inflammation-caused cartilage injury leads to joint dysfunction and pain. Exogenous hyaluronic acid (HA) injection is a well-established treatment, but it has a short duration in vivo and requires multiple injections. Here, a new strategy for in situ reprogramming chondrocytes to continuously produce endogenous high molecular weight HA is developed. This involves a pH-responsive engineered exosome decorated with vesicular stomatitis virus glycoprotein (VSV-G) and hyaluronan synthase type 2 (HAS2). Such engineered exosomes successfully deliver HAS2 to the chondrocyte membranes via VSV-G-mediated membrane fusion triggered by low pH, rather than being degraded in lysosomes. This results in the generation of HAS2-chondrocytes, which are characterized to produce high molecular weight HA in vitro and in vivo. With increased endogenous HA, the injected engineered exosomes enhance cartilage regeneration and inhibit osteoarthritis (OA) progression. Notably, one-shot administration of engineered exosomes drastically increases the intra-articular concentration of high molecular weight HA to 145% of the exogenous HA injection group. Importantly, such endogenous HA is sustained for 4 weeks, whereas the injected exogenous HA rapidly decreases within 2 weeks. The findings demonstrate that pH-responsive engineered exosomes capable of generating endogenous HA hold great potential to replace the treatment of multiple injections of exogenous HA for cartilage repair.
Collapse
Affiliation(s)
- Xiaodan Wu
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Weiyong Tao
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ziyang Lan
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yaping Tian
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhenyu Zhong
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jianwei Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jiaqi Li
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xulong Liu
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xin Zhang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yifan Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jianglin Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Research Base of Regulatory Science for Medical Devices, National Medical Production Administration, Wuhan, 430074, China
- Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bin Zhang
- Department of Orthopedic, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Department of Sports Medicine, Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yingying Du
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Research Base of Regulatory Science for Medical Devices, National Medical Production Administration, Wuhan, 430074, China
- Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Shengmin Zhang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- Research Base of Regulatory Science for Medical Devices, National Medical Production Administration, Wuhan, 430074, China
- Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
8
|
Chen W, Ye Q, Zhang M, Xie R, Xu C. Lubrication for Osteoarthritis: From Single-Function to Multifunctional Lubricants. Int J Mol Sci 2025; 26:1856. [PMID: 40076486 PMCID: PMC11900089 DOI: 10.3390/ijms26051856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that progressively destroys articular cartilage, leading to increased joint friction and severe pain. Therefore, OA can be treated by restoring the lubricating properties of cartilage. In this study, recent advances in lubricants for the treatment of OA are reviewed for both single-function and multifunctional lubricants. Single-function lubricants mainly include glycosaminoglycans, lubricin, and phospholipids, whereas multifunctional lubricants are composed of lubricating and anti-inflammatory bifunctional hydrogels, stem cell-loaded lubricating hydrogels, and drug-loaded lubricating nanoparticles. This review emphasizes the importance of restoring joint lubrication capacity for the treatment of OA and explores the structural features, lubrication properties, and role of these lubricants in modulating intracellular inflammatory responses and metabolism. Current challenges and future research directions in this field are also discussed, with the aim of providing a scientific basis and new ideas for the clinical treatment of OA.
Collapse
Affiliation(s)
- Wen Chen
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China;
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.)
| | - Qianwen Ye
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.)
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Mingshuo Zhang
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.)
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Renjian Xie
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.)
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou 341000, China
| | - Chunming Xu
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
9
|
Erning K, Wilson KL, Smith CS, Nguyen L, Joesph NI, Irengo R, Cao LY, Cumaran M, Shi Y, Lyu S, Riley L, Dunn TW, Carmichael ST, Segura T. Clustered VEGF Nanoparticles in Microporous Annealed Particle (MAP) Hydrogel Accelerates Functional Recovery and Brain Tissue Repair after Stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635733. [PMID: 39974959 PMCID: PMC11838428 DOI: 10.1101/2025.01.30.635733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Ischemic stroke, a blockage in the vasculature of the brain that results in insufficient blood flow, is one of the world's leading causes of disability. The cascade of inflammation and cell death that occurs immediately following stroke drives vascular and functional loss that does not fully recover over time, and no FDA-approved therapies exist that stimulate regeneration post-stroke. We have previously developed a hydrogel scaffold that delivered heparin nanoparticles with and without VEGF bound to their surface to promote angiogenesis and reduce inflammation, respectively. However, the inclusion of the naked heparin nanoparticles warranted concern over the development of bleeding complications. Here, we explore how microporous annealed particle (MAP) scaffolds functionalized with VEGF coated heparin nanoparticles can both reduce inflammation and promote angiogenesis - without the inclusion of free heparin nanoparticles. We show that our updated design not only successfully promotes de novo tissue formation, including the development of mature vessels and neurite sprouting, but it also leads to functional improvement in a photothrombotic stroke model. In addition, we find increased astrocyte infiltration into the infarct site correlated with mature vessel formation. This work demonstrates how our biomaterial design can enhance endogenous regeneration post-stroke while eliminating the need for excess heparin.
Collapse
|
10
|
Keshavarz M, Mohammadi M, Shokrolahi F. Progress in injectable hydrogels for hard tissue regeneration in the last decade. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-39. [PMID: 39853308 DOI: 10.1080/09205063.2024.2436292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/26/2024] [Indexed: 01/26/2025]
Abstract
Bone disorders have increased with increasing the human lifespan, and despite the tissue's ability to self-regeneration, in many congenital problems and hard fractures, bone grafting such as autograft, allograft, and biomaterials implantation through surgery is traditionally used. Because of the adverse effects of these methods, the emergence of injectable hydrogels without the need for surgery and causing more pain for the patient is stunning to develop a new pattern for hard tissue engineering. These materials are formed with various natural and synthetic polymers with a crosslinked network through various chemical methods such as click chemistry, Michael enhancement, Schiff's base and enzymatic reaction and physical interactions with high water absorption which can mimic the environment of cells. The purpose of this research is to review the capabilities of this class of materials in hard tissue regeneration in the last decade through adaptable physical and chemical properties, the ability to fill defect sites with an irregular shape, and the ability to grow hormones or release drugs, in response to external stimuli.
Collapse
Affiliation(s)
- Mahya Keshavarz
- Department of Polymer Engineering, Faculty of Engineering, Qom University of Technology, Qom, Iran
| | - Mohsen Mohammadi
- Department of Polymer Engineering, Faculty of Engineering, Qom University of Technology, Qom, Iran
| | - Fatemeh Shokrolahi
- Department of Biomaterials, Faculty of Science, Iran Polymer and Petrochemical Institute, Tehran, Iran
| |
Collapse
|
11
|
Nasiripour S, Pishbin F, Seyyed Ebrahimi SA. 3D Printing of a Self-Healing, Bioactive, and Dual-Cross-Linked Polysaccharide-Based Composite Hydrogel as a Scaffold for Bone Tissue Engineering. ACS APPLIED BIO MATERIALS 2025; 8:582-599. [PMID: 39764630 DOI: 10.1021/acsabm.4c01476] [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: 01/21/2025]
Abstract
Although 3D printing is becoming a dominant technique for scaffold preparation in bone tissue engineering (TE), developing hydrogel-based ink compositions with bioactive and self-healing properties remains a challenge. This research focuses on developing a bone scaffold based on a composite hydrogel, which maintains its self-healing properties after incorporating bioactive glass and is 3D-printable. The plain hydrogel ink was synthesized using natural polymers of 1 wt % N-carboxyethyl chitosan, 2 wt % hyaluronic acid aldehyde, 0.3 wt % adipic acid hydrazide, and alginate (ALG) (2, 5, and 10 wt %). Bioactive glass (BG) (0 and 5 w/v %) particles were incorporated into the plain matrix to obtain an osteogenic composite hydrogel. The material was characterized via rheology, field emission scanning electron microscopy/energy-dispersive X-ray spectroscopy (FESEM/EDS), Fourier transform infrared (FTIR) spectroscopy, X-ray diffraction (XRD), swelling, degradation, bioactivity, and in vitro cellular assessments. Rheological evaluations confirmed that the specimen with 0 w/v % BG and 5 wt % ALG exhibited the highest G', G″, and viscosity values. All specimens exhibited self-healing, provided by two reversible dynamic bonds, namely, imine and acylhydrazone. Bioactivity evaluation by SBF immersion revealed the formation of HA particles on the composite hydrogels. MTT cytotoxicity assay on MG63 indicated that the composite sample containing 5 w/v % BG and 10 wt % ALG had the highest cell viability (95 ± 1.02%) by culture day 3. The developed approach presents a promising hydrogel ink formulation with a high potential for extrusion-based 3D printing of bone TE constructs.
Collapse
Affiliation(s)
- Saba Nasiripour
- School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, North Kargar Street, Tehran 11155-4563, Iran
| | - Fatemehsadat Pishbin
- School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, North Kargar Street, Tehran 11155-4563, Iran
| | - S A Seyyed Ebrahimi
- Advanced Magnetic Materials Research Center, School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, North Kargar Street, Tehran 11155-4563, Iran
| |
Collapse
|
12
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2025; 14:e2402737. [PMID: 39506433 PMCID: PMC11730424 DOI: 10.1002/adhm.202402737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Wei Liu
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| |
Collapse
|
13
|
Jiang JY, Xue D, Gong JS, Zheng QX, Zhang YS, Su C, Xu ZH, Shi JS. A comprehensive review on microbial hyaluronan-degrading enzymes: from virulence factors to biotechnological tools. BIORESOUR BIOPROCESS 2024; 11:114. [PMID: 39722064 DOI: 10.1186/s40643-024-00832-x] [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: 07/31/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
Hyaluronan (HA), a natural high molecular weight polysaccharide, has extensive applications in cosmetology and medical treatment. Hyaluronan-degrading enzymes (Hyals) act as molecular scissors that cleave HA by breaking the glucosidic linkage. Hyals are present in diverse organisms, including vertebrates, invertebrates and microorganisms, and play momentous roles in biological processes. In recent years, microbial Hyals (mHyals) have gained considerable attention for their exceptional performance in the production and processing of HA. Moreover, the applications of mHyals have been greatly extended to various biomedical fields. To explore the potential applications of mHyals, a thorough comprehension is imperative. In this context, this review systematically summarizes the sources, structures, mechanisms and enzymatic properties of mHyals and discusses their biological functions in host invasion, disease development, and regulation of intestinal flora. Furthermore, versatile applications inspired by their biological functions in medicine development, molecular biology, and industrial biotechnology are comprehensively reviewed. Finally, prospects are presented to emphasize the importance of exploration, expression and characterization of mHyals and the necessity of excavating their potential in biotechnological fields.
Collapse
Affiliation(s)
- Jia-Yu Jiang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China
| | - Dai Xue
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China
- Affiliated Children's Hospital of Jiangnan University, Wuxi, 214023, People's Republic of China
| | - Jin-Song Gong
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China.
| | - Qin-Xin Zheng
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China
| | - Yue-Sheng Zhang
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet 220, 2800, Kgs, Lyngby, Denmark
| | - Chang Su
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China
| | - Zheng-Hong Xu
- School of Biotechnology, Jiangnan University, Wuxi, 214122, People's Republic of China
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, People's Republic of China
| | - Jin-Song Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Lihu Avenue No. 1800, Wuxi, 214122, People's Republic of China.
| |
Collapse
|
14
|
Zhou G, Xu R, Groth T, Wang Y, Yuan X, Ye H, Dou X. The Combination of Bioactive Herbal Compounds with Biomaterials for Regenerative Medicine. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:607-630. [PMID: 38481114 DOI: 10.1089/ten.teb.2024.0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Regenerative medicine aims to restore the function of diseased or damaged tissues and organs by cell therapy, gene therapy, and tissue engineering, along with the adjunctive application of bioactive molecules. Traditional bioactive molecules, such as growth factors and cytokines, have shown great potential in the regulation of cellular and tissue behavior, but have the disadvantages of limited source, high cost, short half-life, and side effects. In recent years, herbal compounds extracted from natural plants/herbs have gained increasing attention. This is not only because herbal compounds are easily obtained, inexpensive, mostly safe, and reliable, but also owing to their excellent effects, including anti-inflammatory, antibacterial, antioxidative, proangiogenic behavior and ability to promote stem cell differentiation. Such effects also play important roles in the processes related to tissue regeneration. Furthermore, the moieties of the herbal compounds can form physical or chemical bonds with the scaffolds, which contributes to improved mechanical strength and stability of the scaffolds. Thus, the incorporation of herbal compounds as bioactive molecules in biomaterials is a promising direction for future regenerative medicine applications. Herein, an overview on the use of bioactive herbal compounds combined with different biomaterial scaffolds for regenerative medicine application is presented. We first introduce the classification, structures, and properties of different herbal bioactive components and then provide a comprehensive survey on the use of bioactive herbal compounds to engineer scaffolds for tissue repair/regeneration of skin, cartilage, bone, neural, and heart tissues. Finally, we highlight the challenges and prospects for the future development of herbal scaffolds toward clinical translation. Overall, it is believed that the combination of bioactive herbal compounds with biomaterials could be a promising perspective for the next generation of regenerative medicine. Impact statement This article reviews the combination of bioactive herbal compounds with biomaterials in the promotion of skin, cartilage, bone, neural, and heart regeneration, due to the anti-inflammatory, antibacterial, antioxidative, and proangiogenic effects of the herbal compounds, but also their effects on the improvement of mechanic strength and stability of biomaterial scaffolds. This review provides a promising direction for the next generation of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Guoying Zhou
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruojiao Xu
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Thomas Groth
- Department of Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Yanying Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xingyu Yuan
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hua Ye
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
- Oxford Suzhou Centre for Advanced Research, University of Oxford, Suzhou, China
| | - Xiaobing Dou
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
15
|
Liu Y, Li S, Zhu J, Fan L, Wang L. Rapid preparation of injectable dual-network hydrogels for biomedical applications using UV-triggered sulfhydryl click reactions. Colloids Surf B Biointerfaces 2024; 244:114180. [PMID: 39217728 DOI: 10.1016/j.colsurfb.2024.114180] [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: 05/09/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The use of hydrogels to mimic natural cartilage implantation can effectively solve the current problems of insufficient cartilage donors and low rate of injury healing. In particular, injectable hydrogels are less invasive in clinical applications and better able to fill uneven injury surfaces. Here, we prepared NorCS and CS-SH by modifying chitosan with 5-norbornene-2-carboxylic acid and N-Acetyl-L-cysteine, respectively. Dual-network hydrogels were prepared by using UV-triggered thiol-ene click reaction between NorCS and CS-SH and the metal coordination between SA and Ca2+. The prepared hydrogels can be cross-linked quickly and exhibit excellent degradability, self-healing and injectable properties. At the same time, the hydrogel also showed good cytocompatibility and could significantly restore the motor function of mice. This study provides an effective strategy for preparing injectable hydrogels capable of rapid cross-linking.
Collapse
Affiliation(s)
- Yanhao Liu
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, PR China
| | - Shubin Li
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, PR China
| | - Jiang Zhu
- Department of Orthopedics, The First Affifiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150001, PR China
| | - Lili Fan
- State Key Laboratory of Black Soils Conservation and Utilization, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun 130102, PR China
| | - Lu Wang
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, PR China.
| |
Collapse
|
16
|
Hridayanka KSN, Duttaroy AK, Basak S. Bioactive Compounds and Their Chondroprotective Effects for Osteoarthritis Amelioration: A Focus on Nanotherapeutic Strategies, Epigenetic Modifications, and Gut Microbiota. Nutrients 2024; 16:3587. [PMID: 39519419 PMCID: PMC11547880 DOI: 10.3390/nu16213587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
In degenerative joint disease like osteoarthritis (OA), bioactive compounds like resveratrol, epigallocatechin gallate, curcumin, and other polyphenols often target various signalling pathways, including NFκB, TGFβ, and Wnt/β-catenin by executing epigenetic-modifying activities. Epigenetic modulation can target genes of disease pathophysiology via histone modification, promoter DNA methylation, and non-coding RNA expression, some of which are directly involved in OA but have been less explored. OA patients often seek options that can improve the quality of their life in addition to existing treatment with nonsteroidal anti-inflammatory drugs (NSAIDs). Although bioactive and natural compounds exhibit therapeutic potential against OA, several disadvantages loom, like insolubility and poor bioavailability. Nanoformulated bioactive compounds promise a better way to alleviate OA since they also control systemic events, including metabolic, immunological, and inflammatory responses, by modulating host gut microbiota that can regulate OA pathogenesis. Recent data suggest gut dysbiosis in OA. However, limited evidence is available on the role of bioactive compounds as epigenetic and gut modulators in ameliorating OA. Moreover, it is not known whether the effects of polyphenolic bioactive compounds on gut microbial response are mediated by epigenetic modulatory activities in OA. This narrative review highlights the nanotherapeutic strategies utilizing bioactive compounds, reporting their effects on chondrocyte growth, metabolism, and epigenetic modifications in osteoarthritis amelioration.
Collapse
Affiliation(s)
- Kota Sri Naga Hridayanka
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway;
| | - Sanjay Basak
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| |
Collapse
|
17
|
German IJS, Barbalho SM, Andreo JC, Zutin TLM, Laurindo LF, Rodrigues VD, Araújo AC, Guiguer EL, Direito R, Pomini KT, Shinohara AL. Exploring the Impact of Catechins on Bone Metabolism: A Comprehensive Review of Current Research and Future Directions. Metabolites 2024; 14:560. [PMID: 39452941 PMCID: PMC11509841 DOI: 10.3390/metabo14100560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/27/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: Degenerative musculoskeletal diseases represent a global health problem due to the progressive deterioration of affected individuals. As a bioactive compound, catechins have shown osteoprotective properties by stimulating osteoblastic cells and inhibiting bone resorption. Thus, this review aimed to address the mechanism of action of catechins on bone tissue. Methods: The search was applied to PubMed without limitations in date, language, or article type. Fifteen articles matched the topic and objective of this review. Results: EGCG (epigallocatechin gallate) and epicatechin demonstrated action on the osteogenic markers RANKL, TRAP, and NF-κβ and expression of BMPs and ALP, thus improving the bone microarchitecture. Studies on animals showed the action of EGCG in increasing calcium and osteoprotegerin levels, in addition to regulating the transcription factor NF-ATc1 associated with osteoclastogenesis. However, it did not show any effect on osteocalcin and RANK. Regarding human studies, EGCG reduced the risk of fracture in a dose-dependent manner. In periodontal tissue, EGCG reduced IL-6, TNF, and RANKL in vitro and in vivo. Human studies showed a reduction in periodontal pockets, gingival index, and clinical attachment level. The action of EGCG on membranes and hydrogels showed biocompatible and osteoinductive properties on the microenvironment of bone tissue by stimulating the expression of osteogenic growth factors and increasing osteocalcin and alkaline phosphate levels, thus promoting new bone formation. Conclusions: EGCG stimulates cytokines related to osteogenes, increasing bone mineral density, reducing osteoclastogenesis factors, and showing great potential as a therapeutic strategy for reducing the risk of bone fractures.
Collapse
Affiliation(s)
- Iris Jasmin Santos German
- Department of Biological Sciences (Anatomy), School of Dentistry of Bauru, University of São Paulo, (FOB-USP), Alameda Doutor Octávio Pinheiro Brisolla, 9-75, Bauru 17012-901, Brazil
| | - Sandra Maria Barbalho
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil (E.L.G.)
- Research Coordination, UNIMAR Charity Hospital, Universidade de Marília (UNIMAR), Marília 17525-902, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, Brazil
| | - Jesus Carlos Andreo
- Department of Biological Sciences (Anatomy), School of Dentistry of Bauru, University of São Paulo, (FOB-USP), Alameda Doutor Octávio Pinheiro Brisolla, 9-75, Bauru 17012-901, Brazil
| | - Tereza Lais Menegucci Zutin
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil (E.L.G.)
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, Brazil; (L.F.L.)
| | - Victória Dogani Rodrigues
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, Brazil; (L.F.L.)
| | - Adriano Cressoni Araújo
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil (E.L.G.)
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, Brazil
| | - Elen Landgraf Guiguer
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil (E.L.G.)
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, Brazil
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed. ULisboa), Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
| | - Karina Torres Pomini
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil (E.L.G.)
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, Brazil
| | - André Luis Shinohara
- Department of Biological Sciences (Anatomy), School of Dentistry of Bauru, University of São Paulo, (FOB-USP), Alameda Doutor Octávio Pinheiro Brisolla, 9-75, Bauru 17012-901, Brazil
| |
Collapse
|
18
|
Nam M, Lee JW, Cha GD. Biomedical Application of Enzymatically Crosslinked Injectable Hydrogels. Gels 2024; 10:640. [PMID: 39451293 PMCID: PMC11507637 DOI: 10.3390/gels10100640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
Hydrogels have garnered significant interest in the biomedical field owing to their tissue-like properties and capability to incorporate various fillers. Among these, injectable hydrogels have been highlighted for their unique advantages, especially their minimally invasive administration mode for implantable use. These injectable hydrogels can be utilized in their pristine forms or as composites by integrating them with therapeutic filler materials. Given their primary application in implantable platforms, enzymatically crosslinked injectable hydrogels have been actively explored due to their excellent biocompatibility and easily controllable mechanical properties for the desired use. This review introduces the crosslinking mechanisms of such hydrogels, focusing on those mediated by horseradish peroxidase (HRP), transglutaminase (TG), and tyrosinase. Furthermore, several parameters and their relationships with the intrinsic properties of hydrogels are investigated. Subsequently, the representative biomedical applications of enzymatically crosslinked-injectable hydrogels are presented, including those for wound healing, preventing post-operative adhesion (POA), and hemostasis. Furthermore, hydrogel composites containing filler materials, such as therapeutic cells, proteins, and drugs, are analyzed. In conclusion, we examine the scientific challenges and directions for future developments in the field of enzymatically crosslinked-injectable hydrogels, focusing on material selection, intrinsic properties, and filler integration.
Collapse
Affiliation(s)
| | | | - Gi Doo Cha
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea; (M.N.); (J.W.L.)
| |
Collapse
|
19
|
Ye H, Zhang R, Zhang C, Xia Y, Jin L. Advances in hyaluronic acid: Bioactivity, complexed biomaterials and biological application: A review. Asian J Surg 2024:S1015-9584(24)01841-4. [PMID: 39217010 DOI: 10.1016/j.asjsur.2024.08.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Hyaluronic acid (HA) is a natural glycosaminoglycan found in the human body, particularly in the extracellular matrix of body fluids and tissues. It plays a critical role in cellular processes of living organisms by maintaining tissue hydration, cell proliferation, differentiation, and inflammatory response. HA exhibits significant biological activity in skin care, aesthetic anti-aging, medical orthopedic repair, gynecological cancer monitoring, and other pathological conditions. Due to its exceptional biocompatibility, biodegradability, lack of toxicity, non-immunogenicity, and its capacity to bond with other substances, various HA-based biomedical products like hydrogels, microneedles, and microspheres have been developed. These innovations have also been applied in various medical and health fields, such as bone and tissue regeneration, gels for medical aesthetic fillers, and gynecology-related cancer treatment, utilizing the HA drug delivery pathway. The interest in HA and its products is increasing due to their biological functions. Therefore, this review aimed to summarize the biological properties of HA and to focus on its applications in the bone tissue engineering and healthcare, for HA has some practical applications of HA-based complexes in biomedical materials, tissue repair, medical aesthetics, and gynecology. Through this review, we seek to offer theoretical research assistance for the development of HA-based bioproducts in the healthcare domain and provide innovative insights for human health.
Collapse
Affiliation(s)
- Huijun Ye
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318 Chaowang Road, Hangzhou, 310005, Zhejiang, China
| | - Ruijuan Zhang
- Center for Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou, 215004, Jiangsu, China
| | - Chunye Zhang
- Center for Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou, 215004, Jiangsu, China
| | - Yujie Xia
- Center for Peak of Excellence on Biological Science and Food Engineering, National University of Singapore (Suzhou) Research Institute, Suzhou, 215004, Jiangsu, China.
| | - Lihua Jin
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318 Chaowang Road, Hangzhou, 310005, Zhejiang, China.
| |
Collapse
|
20
|
Yu Y, Li X, Ying Q, Zhang Z, Liu W, Su J. Synergistic Effects of Shed-Derived Exosomes, Cu 2+, and an Injectable Hyaluronic Acid Hydrogel on Antibacterial, Anti-inflammatory, and Osteogenic Activity for Periodontal Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:33053-33069. [PMID: 38899855 DOI: 10.1021/acsami.4c05062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The primary pathology of periodontitis involves the gradual deterioration of periodontal tissues resulting from the inflammatory reaction triggered by bacterial infection. In this study, a novel drug for periodontal pocket injection, known as the Shed-Cu-HA hydrogel, was developed by incorporating copper ions (Cu2+) and Shed-derived exosomes (Shed-exo) inside the hyaluronic acid (HA) hydrogel. Suitable concentrations of Cu2+ and Shed-exo released from Shed-Cu-HA enhanced cell viability and cell proliferation of human periodontal ligament stem cells. Additionally, the Shed-Cu-HA demonstrated remarkable antibacterial effects against the key periodontal pathogen (Aa) owing to the synergistic effect of Cu2+ and HA. Furthermore, the material effectively suppressed macrophage inflammatory response via the IL-6/JAK2/STAT3 pathway. Moreover, the Shed-Cu-HA, combining the inflammation-regulating properties of HA with the synergistic osteogenic activity of Shed-exo and Cu2+, effectively upregulated the expression of genes and proteins associated with osteogenic differentiation. The experimental findings from a mouse periodontitis model demonstrated that the administration of Shed-Cu-HA effectively reduced the extent of inflammatory cell infiltration and bacterial infections in gingival tissues and facilitated the regeneration of periodontal bone tissues and collagen after 2 and 4 weeks of injection. Consequently, it holds significant prospects for future applications in periodontitis treatment.
Collapse
Affiliation(s)
- Yiqiang Yu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Xuejing Li
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Qiao Ying
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Zhanwei Zhang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Weicai Liu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Jiansheng Su
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| |
Collapse
|
21
|
Kalairaj MS, Pradhan R, Saleem W, Smith MM, Gaharwar AK. Intra-Articular Injectable Biomaterials for Cartilage Repair and Regeneration. Adv Healthc Mater 2024; 13:e2303794. [PMID: 38324655 PMCID: PMC11468459 DOI: 10.1002/adhm.202303794] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/29/2023] [Indexed: 02/09/2024]
Abstract
Osteoarthritis is a degenerative joint disease characterized by cartilage deterioration and subsequent inflammatory changes in the underlying bone. Injectable hydrogels have emerged as a promising approach for controlled drug delivery in cartilage therapies. This review focuses on the latest developments in utilizing injectable hydrogels as vehicles for targeted drug delivery to promote cartilage repair and regeneration. The pathogenesis of osteoarthritis is discussed to provide a comprehensive understanding of the disease progression. Subsequently, the various types of injectable hydrogels used for intra-articular delivery are discussed. Specifically, physically and chemically crosslinked injectable hydrogels are critically analyzed, with an emphasis on their fabrication strategies and their capacity to encapsulate and release therapeutic agents in a controlled manner. Furthermore, the potential of incorporating growth factors, anti-inflammatory drugs, and cells within these injectable hydrogels are discussed. Overall, this review offers a comprehensive guide to navigating the landscape of hydrogel-based therapeutics in osteoarthritis.
Collapse
Affiliation(s)
| | - Ridhi Pradhan
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Waqas Saleem
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Morgan M. Smith
- Department of Veterinary Integrative BiosciencesSchool of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTX77843USA
| | - Akhilesh K. Gaharwar
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Department of Material Science and EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Genetics and Genomics Interdisciplinary ProgramTexas A&M UniversityCollege StationTX77843USA
| |
Collapse
|
22
|
Najihah AZ, Hassan MZ, Ismail Z. Current trend on preparation, characterization and biomedical applications of natural polysaccharide-based nanomaterial reinforcement hydrogels: A review. Int J Biol Macromol 2024; 271:132411. [PMID: 38821798 DOI: 10.1016/j.ijbiomac.2024.132411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/02/2024]
Abstract
The tunable properties of hydrogels have led to their widespread use in various biomedical applications such as wound treatment, drug delivery, contact lenses, tissue engineering and 3D bioprinting. Among these applications, natural polysaccharide-based hydrogels, which are fabricated from materials like agarose, alginate, chitosan, hyaluronic acid, cellulose, pectin and chondroitin sulfate, stand out as preferred choices due to their biocompatibility and advantageous fabrication characteristics. Despite the inherent biocompatibility, polysaccharide-based hydrogels on their own tend to be weak in physiochemical and mechanical properties. Therefore, further reinforcement in the hydrogel is necessary to enhance its suitability for specific applications, ensuring optimal performance in diverse settings. Integrating nanomaterials into hydrogels has proven effective in improving the overall network and performance of the hydrogel. This approach also addresses the limitations associated with pure hydrogels. Next, an overview of recent trends in the fabrication and applications of hydrogels was presented. The characterization of hydrogels was further discussed, focusing specifically on the reinforcement achieved with various hydrogel materials used so far. Finally, a few challenges associated with hydrogels by using polysaccharide-based nanomaterial were also presented.
Collapse
Affiliation(s)
- A Z Najihah
- Faculty of Artificial Intelligence, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100 Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Mohamad Zaki Hassan
- Faculty of Artificial Intelligence, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100 Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| | - Zarini Ismail
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Bandar Baru Nilai, 71800 Nilai, Negeri Sembilan, Malaysia
| |
Collapse
|
23
|
Chang W, Chen L, Chen K. The bioengineering application of hyaluronic acid in tissue regeneration and repair. Int J Biol Macromol 2024; 270:132454. [PMID: 38763255 DOI: 10.1016/j.ijbiomac.2024.132454] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/04/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
The multifaceted role of hyaluronic acid (HA) across diverse biomedical disciplines underscores its versatility in tissue regeneration and repair. HA hydrogels employ different crosslinking including chemical (chitosan, collagen), photo- initiation (riboflavin, LAP), enzymatic (HRP/H2O2), and physical interactions (hydrogen bonds, metal coordination). In biophysics and biochemistry, HA's signaling pathways, primarily through CD44 and RHAMM receptors, modulate cell behavior (cell migration; internalization of HA), inflammation, and wound healing. Particularly, smaller HA fragments stimulate inflammatory responses through toll-like receptors, impacting macrophages and cytokine expression. HA's implications in oncology highlight its involvement in tumor progression, metastasis, and treatment. Elevated HA in tumor stroma impacts apoptosis resistance and promotes tumor growth, presenting potential therapeutic targets to halt tumor progression. In orthopedics, HA's presence in synovial fluid aids in osteoarthritis management, as its supplementation alleviates pain, enhances synovial fluid's viscoelastic properties, and promotes cartilage integrity. In ophthalmology, HA's application in dry eye syndrome addresses symptoms by moisturizing the eyes, replenishing tear film deficiencies, and facilitating wound healing. Intravitreal injections and hydrogel-based systems offer versatile approaches for drug delivery and vitreous humor replacement. For skin regeneration and wound healing, HA hydrogel dressings exhibit exceptional properties by promoting moist wound healing and facilitating tissue repair. Integration of advanced regenerative tools like stem cells and solubilized amnion membranes into HA-based systems accelerates wound closure and tissue recovery. Overall, HA's unique properties and interactions render it a promising candidate across diverse biomedical domains, showcasing immense potentials in tissue regeneration and therapeutic interventions. Nevertheless, many detailed cellular and molecular mechanisms of HA and its applications remain unexplored and warrant further investigation.
Collapse
Affiliation(s)
- WeiTing Chang
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, Taipei, Taiwan
| | - LiRu Chen
- Department of Physical Medicine and Rehabilitation, Mackay Memorial Hospital, Taipei, Taiwan; Department of Mechanical Engineering, National YangMing ChiaoTung University, Hsinchu, Taiwan
| | - KuoHu Chen
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, Taipei, Taiwan; School of Medicine, Tzu-Chi University, Hualien, Taiwan.
| |
Collapse
|
24
|
Pei D, Zeng Z, Geng Z, Cai K, Lu D, Guo C, Guo H, Huang J, Gao B, Yu S. Modulation of macrophage polarization by secondary cross-linked hyaluronan-dopamine hydrogels. Int J Biol Macromol 2024; 270:132417. [PMID: 38759857 DOI: 10.1016/j.ijbiomac.2024.132417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 04/14/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
The inflammatory response plays a critical role in standard tissue repair processes, wherein active modulation of macrophage polarization is necessary for wound healing. Dopamine, a mussel-inspired bioactive material, is widely involved in wound healing, neural/bone/myocardial regeneration, and more. Recent studies indicated that dopamine-modified biomaterials can potentially alter macrophages polarization towards a pro-healing phenotype, thereby enhancing tissue regeneration. Nevertheless the immunoregulatory activity of dopamine on macrophage polarization remains unclear. This study introduces a novel interpenetrating hydrogel to bridge this research gap. The hydrogel, combining varying concentrations of oxidized dopamine with hyaluronic acid hydrogel, allows precise regulation of mechanical properties, antioxidant bioactivity, and biocompatibility. Surprisingly, both in vivo and in vitro outcomes demonstrated that dopamine concentration modulates macrophage polarization, but not linearly. Lower concentration (2 mg/mL) potentially decrease inflammation and facilitate M2 type macrophage polarization. In contrast, higher concentration (10 mg/mL) exhibited a pro-inflammatory tendency in the late stages of implantation. RNA-seq analysis revealed that lower dopamine concentrations induced the M1/M2 transition of macrophages by modulating the NF-κB signaling pathway. Collectively, this research offers valuable insights into the immunoregulation effects of dopamine-integrated biomaterials in tissue repair and regeneration.
Collapse
Affiliation(s)
- Dating Pei
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510500, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China; National Engineering Research Center for Healthcare Devices, Guangzhou 510500, China
| | - Zhiwen Zeng
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510500, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China
| | - Zhijie Geng
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510500, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China; National Engineering Research Center for Healthcare Devices, Guangzhou 510500, China
| | - Kehan Cai
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2008, Australia; National Engineering Research Center for Healthcare Devices, Guangzhou 510500, China
| | - Daohuan Lu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510500, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China; National Engineering Research Center for Healthcare Devices, Guangzhou 510500, China
| | - Cuiping Guo
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510500, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China; National Engineering Research Center for Healthcare Devices, Guangzhou 510500, China
| | - Huilong Guo
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510500, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China
| | - Jun Huang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510500, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China.
| | - Botao Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510500, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China; National Engineering Research Center for Healthcare Devices, Guangzhou 510500, China.
| | - Shan Yu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510500, China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510500, China; National Engineering Research Center for Healthcare Devices, Guangzhou 510500, China.
| |
Collapse
|
25
|
Chapman JH, Ghosh D, Attari S, Ude CC, Laurencin CT. Animal Models of Osteoarthritis: Updated Models and Outcome Measures 2016-2023. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:127-146. [PMID: 38983776 PMCID: PMC11233113 DOI: 10.1007/s40883-023-00309-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2024]
Abstract
Purpose Osteoarthritis (OA) is a global musculoskeletal disorder that affects primarily the knee and hip joints without any FDA-approved disease-modifying therapies. Animal models are essential research tools in developing therapies for OA; many animal studies have provided data for the initiation of human clinical trials. Despite this, there is still a need for strategies to recapitulate the human experience using animal models to better develop treatments and understand pathogenesis. Since our last review on animal models of osteoarthritis in 2016, there have been exciting updates in OA research and models. The main purpose of this review is to update the latest animal models and key features of studies in OA research. Method We used our existing classification method and screened articles in PubMed and bibliographic search for animal OA models between 2016 and 2023. Relevant and high-cited articles were chosen for inclusion in this narrative review. Results Recent studies were analyzed and classified. We also identified ex vivo models as an area of ongoing research. Each animal model offers its own benefit in the study of OA and there are a full range of outcome measures that can be assessed. Despite the vast number of models, each has its drawbacks that have limited translating approved therapies for human use. Conclusion Depending on the outcome measures and objective of the study, researchers should pick the best model for their work. There have been several exciting studies since 2016 that have taken advantage of regenerative engineering techniques to develop therapies and better understand OA. Lay Summary Osteoarthritis (OA) is a chronic debilitating disease without any cure that affects mostly the knee and hip joints and often results in surgical joint replacement. Cartilage protects the joint from mechanical forces and degrades with age or in response to injury. The many contributing causes of OA are still being investigated, and animals are used for preclinical research and to test potential new treatments. A single consensus OA animal model for preclinical studies is non-existent. In this article, we review the many animal models for OA and provide a much-needed update on studies and model development since 2016.
Collapse
Affiliation(s)
- James H. Chapman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Seyyedmorteza Attari
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Chinedu C. Ude
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical and Bimolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
26
|
Nie R, Zhang QY, Feng ZY, Huang K, Zou CY, Fan MH, Zhang YQ, Zhang JY, Li-Ling J, Tan B, Xie HQ. Hydrogel-based immunoregulation of macrophages for tissue repair and regeneration. Int J Biol Macromol 2024; 268:131643. [PMID: 38643918 DOI: 10.1016/j.ijbiomac.2024.131643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 04/23/2024]
Abstract
The rational design of hydrogel materials to modulate the immune microenvironment has emerged as a pivotal approach in expediting tissue repair and regeneration. Within the immune microenvironment, an array of immune cells exists, with macrophages gaining prominence in the field of tissue repair and regeneration due to their roles in cytokine regulation to promote regeneration, maintain tissue homeostasis, and facilitate repair. Macrophages can be categorized into two types: classically activated M1 (pro-inflammatory) and alternatively activated M2 (anti-inflammatory and pro-repair). By regulating the physical and chemical properties of hydrogels, the phenotypic transformation and cell behavior of macrophages can be effectively controlled, thereby promoting tissue regeneration and repair. A full understanding of the interaction between hydrogels and macrophages can provide new ideas and methods for future tissue engineering and clinical treatment. Therefore, this paper reviews the effects of hydrogel components, hardness, pore size, and surface morphology on cell behaviors such as macrophage proliferation, migration, and phenotypic polarization, and explores the application of hydrogels based on macrophage immune regulation in skin, bone, cartilage, and nerve tissue repair. Finally, the challenges and future prospects of macrophage-based immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Qing-Yi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Zi-Yuan Feng
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Kai Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Chen-Yu Zou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ming-Hui Fan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yue-Qi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ji-Ye Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Medical Genetics, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Bo Tan
- Department of Orthopedic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, PR China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, PR China.
| |
Collapse
|
27
|
Han J, Choi S, Hong J, Gang D, Lee S, Shin K, Ko J, Kim JU, Hwang NS, An YH, Gu M, Kim SH. Superoxide Dismutase-Mimetic Polyphenol-Based Carbon Dots for Multimodal Bioimaging and Treatment of Atopic Dermatitis. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38686704 DOI: 10.1021/acsami.4c02634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Polyphenols have been investigated for their potential to mitigate inflammation in the context of atopic dermatitis (AD). In this study, epigallocatechin-3-gallate (EGCG)-based carbon dots (EGCG@CDs) were developed to enhance transdermal penetration, reduce inflammation, recapitulate superoxide dismutase (SOD) activity, and provide antimicrobial effects for AD treatment. The water-soluble EGCG@CDs in a few nanometers size exhibit a negative zeta potential, making them suitable for effective transdermal penetration. The fluorescence properties, including an upconversion effect, make EGCG@CDs suitable imaging probes for both in vitro and in vivo applications. By mimicking the SOD enzyme, EGCG@CDs scavenge reactive oxygen species (ROS) and actively produce hydrogen peroxide through a highly catalytic capability toward the oxygen reduction reaction, resulting in the inhibition of bacterial growth. The enhanced antioxidant properties, high charge mobility, and various functional groups of EGCG@CDs prove effective in reducing intracellular ROS in an in vitro AD model. In the mouse AD model, EGCG@CDs incorporated into a hydrogel actively penetrated the epidermal layer, leading to ROS scavenging, reduced mast cell activation, and histological recovery of skin barriers. This research represents the versatile potential of EGCG@CDs in addressing AD and advancing tissue engineering.
Collapse
Affiliation(s)
- Jeongmin Han
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
| | - Sumi Choi
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
| | - Jinwoo Hong
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
| | - Dayeong Gang
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
| | - Seunghoon Lee
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
- Department of Chemistry, Dong-A University, Busan 49315, Republic of Korea
| | - Kwangsoo Shin
- Department of Polymer Science and Engineering and Program in Environmental and Polymer Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Junghyeon Ko
- School of Chemical and Biological Engineering, The Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong-Uk Kim
- School of Chemical and Biological Engineering, The Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, The Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Bio-MAX/N-Bio, Institute of Bioengineering, Institute of Engineering Research, Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Hyeon An
- School of Chemical and Biological Engineering, The Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Bio-MAX/N-Bio, Institute of Bioengineering, Institute of Engineering Research, Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Minsu Gu
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
| |
Collapse
|
28
|
Chandel AKS, Sreedevi Madhavikutty A, Okada S, Qiming Z, Inagaki NF, Ohta S, Ito T. Injectable, shear-thinning, photocrosslinkable, and tissue-adhesive hydrogels composed of diazirine-modified hyaluronan and dendritic polyethyleneimine. Biomater Sci 2024; 12:1454-1464. [PMID: 38223981 DOI: 10.1039/d3bm01279d] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
In the present study, we report the first synthesis of diazirine-modified hyaluronic acid (HA-DAZ). In addition, we also produced a precursor polymer solution composed of HA-DAZ and dendritic polyethyleneimine (DPI) that showed strong shear-thinning properties. Furthermore, its viscosity was strongly reduced (i.e., from 5 × 105 mPa s at 10-3 s-1 to 6 × 101 mPa s at 103 s-1), substantially, which enhanced solution injectability using a 21 G needle. After ultraviolet irradiation at 365 nm and 6 mW cm-2, the HA-DAZ/DPI solution achieved rapid gelation, as measured using the stirring method, and its gelation time decreased from 200 s to 9 s as the total concentrations of HA-DAZ and DPI increased. Following UV irradiation, the storage modulus increased from 40 to 200 Pa. In addition, reversible sol-gel transition and self-healing properties were observed even after UV irradiation. This suggests that the HA-DAZ/DPI hydrogel was crosslinked in multiple ways, i.e., via covalent bonding between the diazirine and amine groups and via intermolecular interactions, including hydrogen bonding, electrostatic interactions, and hydrophobic interactions. A lap shear test showed that the HA-DAZ/DPI hydrogel exhibited strong adhesiveness as a fibrin glue following UV irradiation. Finally, the HA-DAZ/DPI hydrogel showed higher tissue reinforcement than fibrin glue in an ex vivo burst pressure test of the porcine esophageal mucosa.
Collapse
Affiliation(s)
- Arvind K Singh Chandel
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Athira Sreedevi Madhavikutty
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Saki Okada
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Zhang Qiming
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Natsuko F Inagaki
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Seiichi Ohta
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Taichi Ito
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Radiology and Biomedical Engineering, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
29
|
Kamedani M, Okawa M, Madhavikutty AS, Tsai CC, Singh Chandel AK, Fujiyabu T, Inagaki NF, Ito T. Injectable Extracellular Matrix-Inspired Hemostatic Hydrogel Composed of Hyaluronan and Gelatin with Shear-Thinning and Self-Healing. Biomacromolecules 2024; 25:1790-1799. [PMID: 38306215 DOI: 10.1021/acs.biomac.3c01251] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Abstract
Injectable ECM-inspired hydrogels composed of hyaluronic acid and gelatin are biocompatible and potentially useful for various medical applications. We developed injectable hydrogels composed of monoaldehyde-modified hyaluronic acid (HA-mCHO) and carbohydrazide-modified gelatin (GL-CDH), "HA/GL gel", whose ratios of HA-mCHO to GL-CDH were different. The hydrogels exhibited gelation times shorter than 3 s. In addition, the hydrogels showed strong shear-thinning and self-healing properties, mainly because of the dynamic covalent bonding of Schiff bases between HA-mCHO and GL-CDH. This hydrogel degraded in the mice's peritoneum for a week and showed excellent biocompatibility. Moreover, the hydrogel showed a higher breaking strength than fibrin glue in the lap shear test of porcine skin. Finally, the hydrogels decreased bleeding to as low as fibrin glue without using thrombin and fibrinogen in a mouse liver bleeding model in both single- and double-barreled syringe administrations. HA/GL gels have the potential for excellent biocompatibility and hemostasis in clinical settings.
Collapse
Affiliation(s)
- Momoko Kamedani
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Masashi Okawa
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Athira Sreedevi Madhavikutty
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Ching-Cheng Tsai
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Arvind K Singh Chandel
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takeshi Fujiyabu
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Natsuko F Inagaki
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Taichi Ito
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
30
|
Hu Y, Wang Y, Yang F, Liu D, Lu G, Li S, Wei Z, Shen X, Jiang Z, Zhao Y, Pang Q, Song B, Shi Z, Shafique S, Zhou K, Chen X, Su W, Jian J, Tang K, Liu T, Zhu Y. Flexible Organic Photovoltaic-Powered Hydrogel Bioelectronic Dressing With Biomimetic Electrical Stimulation for Healing Infected Diabetic Wounds. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307746. [PMID: 38145346 PMCID: PMC10933690 DOI: 10.1002/advs.202307746] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/28/2023] [Indexed: 12/26/2023]
Abstract
Electrical stimulation (ES) is proposed as a therapeutic solution for managing chronic wounds. However, its widespread clinical adoption is limited by the requirement of additional extracorporeal devices to power ES-based wound dressings. In this study, a novel sandwich-structured photovoltaic microcurrent hydrogel dressing (PMH dressing) is designed for treating diabetic wounds. This innovative dressing comprises flexible organic photovoltaic (OPV) cells, a flexible micro-electro-mechanical systems (MEMS) electrode, and a multifunctional hydrogel serving as an electrode-tissue interface. The PMH dressing is engineered to administer ES, mimicking the physiological injury current occurring naturally in wounds when exposed to light; thus, facilitating wound healing. In vitro experiments are performed to validate the PMH dressing's exceptional biocompatibility and robust antibacterial properties. In vivo experiments and proteomic analysis reveal that the proposed PMH dressing significantly accelerates the healing of infected diabetic wounds by enhancing extracellular matrix regeneration, eliminating bacteria, regulating inflammatory responses, and modulating vascular functions. Therefore, the PMH dressing is a potent, versatile, and effective solution for diabetic wound care, paving the way for advancements in wireless ES wound dressings.
Collapse
Affiliation(s)
- Yi‐Wei Hu
- Health Science CenterNingbo UniversityNingbo315211P. R. China
- Orthopaedic Oncology Center of Changzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Yu‐Heng Wang
- Faculty of Electrical Engineering and Computer ScienceNingbo UniversityNingbo315211P. R. China
- State Key Laboratory of Electrical Insulation and Power EquipmentXi'an Jiaotong UniversityXi'an710049P. R. China
- CAS Key Laboratory of Nanosystem and Hierarchical FabricationNational Center for Nanoscience and TechnologyUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Fang Yang
- Health Science CenterNingbo UniversityNingbo315211P. R. China
| | - Ding‐Xin Liu
- State Key Laboratory of Electrical Insulation and Power EquipmentXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Guang‐Hao Lu
- State Key Laboratory of Electrical Insulation and Power EquipmentXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Sheng‐Tao Li
- State Key Laboratory of Electrical Insulation and Power EquipmentXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Zhi‐Xiang Wei
- CAS Key Laboratory of Nanosystem and Hierarchical FabricationNational Center for Nanoscience and TechnologyUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Xiang Shen
- The Research Institute of Advanced TechnologiesNingbo UniversityNingbo315211P. R. China
| | - Zhuang‐De Jiang
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Yi‐Fan Zhao
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Qian Pang
- Health Science CenterNingbo UniversityNingbo315211P. R. China
| | - Bai‐Yang Song
- Health Science CenterNingbo UniversityNingbo315211P. R. China
| | - Ze‐Wen Shi
- Health Science CenterNingbo UniversityNingbo315211P. R. China
| | - Shareen Shafique
- School of Physical Science and TechnologyNingbo UniversityNingbo315211P. R. China
| | - Kun Zhou
- Shenzhen Institute of Aggregate Science and TechnologyThe Chinese University of Hong Kong ShenzhenShenzhen518172P. R. China
| | - Xiao‐Lian Chen
- Printable Electronics Research Center & Nano‐Device and Materials DivisionSuzhou Institute of Nano‐Tech and Nano‐BionicsNano Chinese Academy of SciencesSuzhou215123P. R. China
| | - Wen‐Ming Su
- Printable Electronics Research Center & Nano‐Device and Materials DivisionSuzhou Institute of Nano‐Tech and Nano‐BionicsNano Chinese Academy of SciencesSuzhou215123P. R. China
| | - Jia‐Wen Jian
- Faculty of Electrical Engineering and Computer ScienceNingbo UniversityNingbo315211P. R. China
| | - Ke‐Qi Tang
- Institute of Mass SpectrometrySchool of Material Science and Chemical EngineeringNingbo UniversityNingbo315211P. R. China
| | - Tie‐Long Liu
- Orthopaedic Oncology Center of Changzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Ya‐Bin Zhu
- Health Science CenterNingbo UniversityNingbo315211P. R. China
| |
Collapse
|
31
|
Cai R, Shan Y, Du F, Miao Z, Zhu L, Hang L, Xiao L, Wang Z. Injectable hydrogels as promising in situ therapeutic platform for cartilage tissue engineering. Int J Biol Macromol 2024; 261:129537. [PMID: 38278383 DOI: 10.1016/j.ijbiomac.2024.129537] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/01/2024] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
Injectable hydrogels are gaining prominence as a biocompatible, minimally invasive, and adaptable platform for cartilage tissue engineering. Commencing with their synthesis, this review accentuates the tailored matrix formulations and cross-linking techniques essential for fostering three-dimensional cell culture and melding with complex tissue structures. Subsequently, it spotlights the hydrogels' enhanced properties, highlighting their augmented functionalities and broadened scope in cartilage tissue repair applications. Furthermore, future perspectives are advocated, urging continuous innovation and exploration to surmount existing challenges and harness the full clinical potential of hydrogels in regenerative medicine. Such advancements are crucial for validating the long-term efficacy and safety of hydrogels, positioning them as a promising direction in regenerative medicine to address cartilage-related ailments.
Collapse
Affiliation(s)
- Rong Cai
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Yisi Shan
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Fengyi Du
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212013, China
| | - Zhiwei Miao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Like Zhu
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Li Hang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Long Xiao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China.
| | - Zhirong Wang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China.
| |
Collapse
|
32
|
Wu M, Zheng K, Li W, He W, Qian C, Lin Z, Xiao H, Yang H, Xu Y, Wei M, Bai J, Geng D. Nature‐Inspired Strategies for the Treatment of Osteoarthritis. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202305603] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Indexed: 01/06/2025]
Abstract
AbstractArticular cartilage is devoid of nerves and blood vessels, and its nutrients must be obtained from the joint fluid; therefore, its ability to repair itself is limited. Manufactured materials such as artificial cartilage or synthetic materials are typically used in traditional approaches for knee cartilage repair. However, durability, postimplant rejection, and tissue incompatibility are the problems associated with these materials. In recent decades, tissue engineering and regenerative medicine have focused on the development of functional substitutes, particularly those based on naturally inspired biopolymers. This review focuses on sustainably produced biopolymers based on materials derived from natural sources. Furthermore, these materials have many advantages, including low antigenicity, biocompatibility, and degradability. Of course, there are also many challenges associated with natural materials, such as the lack of clinical studies and long‐term follow‐up data, unstable mechanical properties of the materials, and high demands placed on preparation and molding techniques. In this review, an overview of natural and nature‐inspired polymers that are the subject of research to date, as well as their structural designs and product performances is provided. This review provides scientific guidance for enhancing the development of naturally inspired materials for treating cartilage injuries.
Collapse
Affiliation(s)
- Mingzhou Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University No. 188 Shizi Road Suzhou Jiangsu 215006 China
- Department of Orthopedics Taicang TCM Hospital Affiliated to Nanjing University of Chinese Medicine No. 140 Renmin South Road Suzhou Jiangsu 215400 China
| | - Kai Zheng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University No. 188 Shizi Road Suzhou Jiangsu 215006 China
| | - Wenhao Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University No. 188 Shizi Road Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China Hefei Anhui 230031 China
- National Center for Translational Medicine (Shanghai) SHU Branch Shanghai University Shanghai 215031 China
| | - Weiming He
- Affiliated Hospital of Nanjing University of Chinese Medicine Nanjing Jiangsu 210004 China
| | - Chen Qian
- Department of Orthopedics, The First Affiliated Hospital of Soochow University No. 188 Shizi Road Suzhou Jiangsu 215006 China
| | - Zhixiang Lin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University No. 188 Shizi Road Suzhou Jiangsu 215006 China
| | - Haixiang Xiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University No. 188 Shizi Road Suzhou Jiangsu 215006 China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University No. 188 Shizi Road Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University No. 188 Shizi Road Suzhou Jiangsu 215006 China
| | - Minggang Wei
- Department of Traditional Chinese Medicine The First Affiliated Hospital of Soochow University Suzhou Jiangsu 215006 China
| | - Jiaxiang Bai
- Department of Orthopedics The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China Hefei Anhui 230031 China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University No. 188 Shizi Road Suzhou Jiangsu 215006 China
| |
Collapse
|
33
|
Zhao D, Cheng Q, Geng H, Liu J, Zhang Y, Cui J, Liu C, Cheng L. Decoding Macrophage Subtypes to Engineer Modulating Hydrogels for the Alleviation of Intervertebral Disk Degeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304480. [PMID: 37939288 PMCID: PMC10767410 DOI: 10.1002/advs.202304480] [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: 07/04/2023] [Revised: 09/26/2023] [Indexed: 11/10/2023]
Abstract
A major pathological basis for low back pain is intervertebral disk degeneration, which is primarily caused by the degeneration of nucleus pulposus cells due to imbalances in extracellular matrix (ECM) anabolism and catabolism. The phenotype of macrophages in the local immune microenvironment greatly influences the balance of ECM metabolism. Therefore, the control over the macrophage phenotype of the ECM is promising to repair intervertebral disk degeneration. Herein, the preparation of an injectable nanocomposite hydrogel is reported by embedding epigallocatechin-3-gallate-coated hydroxyapatite nanorods in O-carboxymethyl chitosan cross-linked with aldehyde hyaluronic acid that is capable of modulating the phenotype of macrophages. The bioactive components play a primary role in repairing the nucleus pulposus, where the hydroxyapatite nanorods can promote anabolism in the ECM through the nucleopulpogenic differentiation of mesenchymal stem cells. In addition, epigallocatechin-3-gallate can decrease catabolism in the ECM in nucleus pulposus by inducing M2 macrophage polarization, which exists in normal intervertebral disks and can alleviate degeneration. The nanocomposite hydrogel system shows promise for the minimally invasive and effective treatment of intervertebral disk degeneration by controlling anabolism and catabolism in the ECM and inhibiting the IL17 signaling pathway (M1-related pathway) in vitro and in vivo.
Collapse
Affiliation(s)
- Da‐Wang Zhao
- Department of OrthopedicsQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Qian Cheng
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of EducationSchool of Chemistry and Chemical EngineeringShandong UniversityJinanShandong250100China
| | - Huimin Geng
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of EducationSchool of Chemistry and Chemical EngineeringShandong UniversityJinanShandong250100China
| | - Jinbo Liu
- Department of OrthopedicsQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Yuanqiang Zhang
- Department of OrthopedicsQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of EducationSchool of Chemistry and Chemical EngineeringShandong UniversityJinanShandong250100China
| | - Chao Liu
- Department of Oral and Maxillofacial SurgeryQilu Hospital of Shandong UniversityJinanShandong250012China
- Department of Oral Surgery, Shanghai Key Laboratory of StomatologyNational Clinical Research Center of StomatologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Lei Cheng
- Department of OrthopedicsQilu Hospital of Shandong UniversityJinanShandong250012China
| |
Collapse
|
34
|
Mohanto S, Narayana S, Merai KP, Kumar JA, Bhunia A, Hani U, Al Fatease A, Gowda BHJ, Nag S, Ahmed MG, Paul K, Vora LK. Advancements in gelatin-based hydrogel systems for biomedical applications: A state-of-the-art review. Int J Biol Macromol 2023; 253:127143. [PMID: 37793512 DOI: 10.1016/j.ijbiomac.2023.127143] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023]
Abstract
A gelatin-based hydrogel system is a stimulus-responsive, biocompatible, and biodegradable polymeric system with solid-like rheology that entangles moisture in its porous network that gradually protrudes to assemble a hierarchical crosslinked arrangement. The hydrolysis of collagen directs gelatin construction, which retains arginyl glycyl aspartic acid and matrix metalloproteinase-sensitive degeneration sites, further confining access to chemicals entangled within the gel (e.g., cell encapsulation), modulating the release of encapsulated payloads and providing mechanical signals to the adjoining cells. The utilization of various types of functional tunable biopolymers as scaffold materials in hydrogels has become highly attractive due to their higher porosity and mechanical ability; thus, higher loading of proteins, peptides, therapeutic molecules, etc., can be further modulated. Furthermore, a stimulus-mediated gelatin-based hydrogel with an impaired concentration of gellan demonstrated great shear thinning and self-recovering characteristics in biomedical and tissue engineering applications. Therefore, this contemporary review presents a concise version of the gelatin-based hydrogel as a conceivable biomaterial for various biomedical applications. In addition, the article has recapped the multiple sources of gelatin and their structural characteristics concerning stimulating hydrogel development and delivery approaches of therapeutic molecules (e.g., proteins, peptides, genes, drugs, etc.), existing challenges, and overcoming designs, particularly from drug delivery perspectives.
Collapse
Affiliation(s)
- Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India.
| | - Soumya Narayana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Khushboo Paresh Merai
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Jahanvee Ashok Kumar
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Adrija Bhunia
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - B H Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India; School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast BT9 7BL, UK.
| | - Sagnik Nag
- Department of Bio-Sciences, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Tiruvalam Rd, 632014, Tamil Nadu, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Karthika Paul
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast BT9 7BL, UK
| |
Collapse
|
35
|
Hu X, Wu H, Yong X, Wang Y, Yang S, Fan D, Xiao Y, Che L, Shi K, Li K, Xiong C, Zhu H, Qian Z. Cyclical endometrial repair and regeneration: Molecular mechanisms, diseases, and therapeutic interventions. MedComm (Beijing) 2023; 4:e425. [PMID: 38045828 PMCID: PMC10691302 DOI: 10.1002/mco2.425] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 10/21/2023] [Accepted: 10/27/2023] [Indexed: 12/05/2023] Open
Abstract
The endometrium is a unique human tissue with an extraordinary ability to undergo a hormone-regulated cycle encompassing shedding, bleeding, scarless repair, and regeneration throughout the female reproductive cycle. The cyclical repair and regeneration of the endometrium manifest as changes in endometrial epithelialization, glandular regeneration, and vascularization. The mechanisms encompass inflammation, coagulation, and fibrinolytic system balance. However, specific conditions such as endometriosis or TCRA treatment can disrupt the process of cyclical endometrial repair and regeneration. There is uncertainty about traditional clinical treatments' efficacy and side effects, and finding new therapeutic interventions is essential. Researchers have made substantial progress in the perspective of regenerative medicine toward maintaining cyclical endometrial repair and regeneration in recent years. Such progress encompasses the integration of biomaterials, tissue-engineered scaffolds, stem cell therapies, and 3D printing. This review analyzes the mechanisms, diseases, and interventions associated with cyclical endometrial repair and regeneration. The review discusses the advantages and disadvantages of the regenerative interventions currently employed in clinical practice. Additionally, it highlights the significant advantages of regenerative medicine in this domain. Finally, we review stem cells and biologics among the available interventions in regenerative medicine, providing insights into future therapeutic strategies.
Collapse
Affiliation(s)
- Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu UniversityChengdu UniversityChengduSichuanChina
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Haoming Wu
- Clinical Medical College and Affiliated Hospital of Chengdu UniversityChengdu UniversityChengduSichuanChina
| | - Xin Yong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of BiotherapySichuan UniversityChengduSichuanChina
| | - Yao Wang
- Clinical Medical College and Affiliated Hospital of Chengdu UniversityChengdu UniversityChengduSichuanChina
| | - Shuhao Yang
- Department of OrthopedicsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Diyi Fan
- Clinical Medical College and Affiliated Hospital of Chengdu UniversityChengdu UniversityChengduSichuanChina
| | - Yibo Xiao
- Clinical Medical College and Affiliated Hospital of Chengdu UniversityChengdu UniversityChengduSichuanChina
| | - Lanyu Che
- Clinical Medical College and Affiliated Hospital of Chengdu UniversityChengdu UniversityChengduSichuanChina
| | - Kun Shi
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Kainan Li
- Clinical Medical College and Affiliated Hospital of Chengdu UniversityChengdu UniversityChengduSichuanChina
| | | | - Huili Zhu
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of EducationWest China Second University Hospital of Sichuan UniversityChengduSichuanChina
| | - Zhiyong Qian
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
36
|
Xu Y, Wang JY, Meng T, Ma XW, Li H, Li K. Role of hydrogels in osteoarthritis: A comprehensive review. Int J Rheum Dis 2023; 26:2390-2401. [PMID: 37934919 DOI: 10.1111/1756-185x.14968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023]
Abstract
Osteoarthritis (OA) is a chronic, degenerative, and age-related disease. It is characterized by chronic inflammation, progressive articular cartilage destruction, and subchondral bone sclerosis. The current effective treatment for OA is limited. Hydrogel is a kind of unique carrier with well-known biocompatibility, softness, and high water content among various biomaterials. Hydrogels are developed for different biomedical applications, for instance, drug delivery, and tissue engineering. To date, a variety of hydrogels-based therapies have been used in OA patients or animal models. In this review, we comprehensively summarized the potential role of hydrogels in chondrocytes proliferation, apoptosis, and inflammatory component production and discussed the impact of hydrogels on OA development. The collection of this information will help better understand the present progress of hydrogels in OA.
Collapse
Affiliation(s)
- Yuan Xu
- College of Health Industry, Sichuan Tourism University, Chengdu, China
| | - Jing-Yan Wang
- College of Health Industry, Sichuan Tourism University, Chengdu, China
| | - Tian Meng
- College of Health Industry, Sichuan Tourism University, Chengdu, China
| | - Xue-Wei Ma
- College of Health Industry, Sichuan Tourism University, Chengdu, China
| | - Hao Li
- College of Health Industry, Sichuan Tourism University, Chengdu, China
| | - Kai Li
- College of Health Industry, Sichuan Tourism University, Chengdu, China
| |
Collapse
|
37
|
Khan NM, Uddin M, Falade EO, Khan FA, Wang J, Shafique M, Alnemari RM, Abduljabbar MH, Ahmad S. Green Synthesis of Low-Glycemic Amylose-Lipid Nanocomposites by High-Speed Homogenization and Formulation into Hydrogel. Molecules 2023; 28:7154. [PMID: 37894632 PMCID: PMC10608987 DOI: 10.3390/molecules28207154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
In this research, we focused on the production of amylose-lipid nanocomposite material (ALN) through a green synthesis technique utilizing high-speed homogenization. Our aim was to investigate this novel material's distinctive physicochemical features and its potential applications as a low-glycemic gelling and functional food ingredient. The study begins with the formulation of the amylose-lipid nanomaterial from starch and fatty acid complexes, including stearic, palmitic, and lauric acids. Structural analysis reveals the presence of ester carbonyl functionalities, solid matrix structures, partial crystallinities, and remarkable thermal stability within the ALN. Notably, the ALN exhibits a significantly low glycemic index (GI, 40%) and elevated resistance starch (RS) values. The research extends to the formulation of ALN into nanocomposite hydrogels, enabling the evaluation of its anthocyanin absorption capacity. This analysis provides valuable insights into the rheological properties and viscoelastic behavior of the resulting hydrogels. Furthermore, the study investigates anthocyanin encapsulation and retention by ALN-based hydrogels, with a particular focus on the influence of pH and physical cross-link networks on the uptake capacity presenting stearic-acid (SA) hydrogel with the best absorption capacity. In conclusion, the green-synthesized (ALN) shows remarkable functional and structural properties. The produced ALN-based hydrogels are promising materials for a variety of applications, such as medicine administration, food packaging, and other industrial purposes.
Collapse
Affiliation(s)
- Nasir Mehmood Khan
- Department of Agriculture, Shaheed Benazir Bhutto University, Sheringal, Upper Dir 18000, Khyber Pakhtunkhwa, Pakistan
| | - Misbah Uddin
- Department of Chemistry, Shaheed Benazir Bhutto University, Sheringal, Upper Dir 18000, Khyber Pakhtunkhwa, Pakistan
| | - Ebenezer Ola Falade
- Institute of Food Science and Technology, Chinese Academy of Agriculture Sciences, Beijing 100193, China
| | - Farman Ali Khan
- Department of Chemistry, Shaheed Benazir Bhutto University, Sheringal, Upper Dir 18000, Khyber Pakhtunkhwa, Pakistan
| | - Jian Wang
- Department of Chinese Materia Medica, Chongqing College of Traditional Chinese Medicine, Chongqing 402760, China
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Muhammad Shafique
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 15571, Saudi Arabia
| | - Reem M Alnemari
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Maram H Abduljabbar
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Shujaat Ahmad
- Department of Pharmacy, Shaheed Benazir Bhutto University, Sheringal, Upper Dir 18000, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
38
|
Jiang Y, Liao H, Yan L, Jiang S, Zheng Y, Zhang X, Wang K, Wang Q, Han L, Lu X. A Metal-Organic Framework-Incorporated Hydrogel for Delivery of Immunomodulatory Neobavaisoflavone to Promote Cartilage Regeneration in Osteoarthritis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:46598-46612. [PMID: 37769191 DOI: 10.1021/acsami.3c06706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
The treatment of osteoarthritis (OA)-related cartilage defects is a great clinical challenge due to the complex pathogenesis of OA and poor self-repair ability of cartilage tissue. Combining local and long-term anti-inflammatory therapies to promote cartilage repair is an effective method to treat OA. In this study, a zinc-organic framework-incorporated extracellular matrix (ECM)-mimicking hydrogel platform was constructed for the inflammatory microenvironment-responsive delivery of neobavaisoflavone (NBIF) to promote cartilage regeneration in OA. The NBIF was encapsulated in situ in zeolitic imidazolate frameworks (ZIF-8 MOFs). The NBIF@ZIF-8 MOFs were decorated with polydopamine and incorporated into a methacrylate gelatin/hyaluronic acid hybrid network to form the NBIF@ZIF-8/PHG hydrogel. The hydrogel featured excellent cell/tissue affinity, providing a favorable microenvironment for recruiting cells and cytokines to the defect sites. The hydrogel enabled the on-demand NBIF released in response to a weakly acidic microenvironment at the injured joint site to resolve inflammatory responses during the early stages of OA. Consequently, the cooperativity of the loaded NBIF and hydrogel synergistically modulated the immune response and assisted in cartilage defect repair. In summary, the NBIF@ZIF-8/PHG hydrogel delivery platform represents an effective treatment strategy for OA-related cartilage defects and may attract attentions for applications in other inflammatory diseases.
Collapse
Affiliation(s)
- Yanan Jiang
- Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Haixia Liao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Liwei Yan
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Shengxi Jiang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Yujia Zheng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Xin Zhang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Kefeng Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610064, China
| | - Lu Han
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, School of Medicine and Pharmaceutics, Ocean University of China, Qingdao, Shandong 266003, China
| | - Xiong Lu
- Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| |
Collapse
|
39
|
Su J, Yu M, Wang H, Wei Y. Natural anti-inflammatory products for osteoarthritis: From molecular mechanism to drug delivery systems and clinical trials. Phytother Res 2023; 37:4321-4352. [PMID: 37641442 DOI: 10.1002/ptr.7935] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 08/31/2023]
Abstract
Osteoarthritis (OA) is a degenerative joint disease that affects millions globally. The present nonsteroidal anti-inflammatory drug treatments have different side effects, leading researchers to focus on natural anti-inflammatory products (NAIPs). To review the effectiveness and mechanisms of NAIPs in the cellular microenvironment, examining their impact on OA cell phenotype and organelles levels. Additionally, we summarize relevant research on drug delivery systems and clinical randomized controlled trials (RCTs), to promote clinical studies and explore natural product delivery options. English-language articles were searched on PubMed using the search terms "natural products," "OA," and so forth. We categorized search results based on PubChem and excluded "natural products" which are mix of ingredients or compounds without the structure message. Then further review was separately conducted for molecular mechanisms, drug delivery systems, and RCTs later. At present, it cannot be considered that NAIPs can thoroughly prevent or cure OA. Further high-quality studies on the anti-inflammatory mechanism and drug delivery systems of NAIPs are needed, to determine the appropriate drug types and regimens for clinical application, and to explore the combined effects of different NAIPs to prevent and treat OA.
Collapse
Affiliation(s)
- Jianbang Su
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Minghao Yu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haochen Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yingliang Wei
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
40
|
Li Y, Chen X, Zhou Z, Fang B, Chen Z, Huang Y, Hu Y, Liu H. Berberine oleanolic acid complex salt grafted hyaluronic acid/silk fibroin (BOA-g-HA/SF) composite scaffold promotes cartilage tissue regeneration under IL-1β caused stress. Int J Biol Macromol 2023; 250:126104. [PMID: 37536412 DOI: 10.1016/j.ijbiomac.2023.126104] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Since inflammatory cytokines cause stress to chondrocytes and the failure of cartilage defects repair with cartilage tissue engineering, it is necessary to develop a scaffold to maintain cartilage regeneration under inflammatory factors caused stress. Following a berberine-oleanolic acid (OA) complex salt (BOA) was grafted to hyaluronic acid (HA) to obtain water soluble BOA-g-HA, it mixed with silk fibroin (SF) to prepared 4 solutions, which contained 30 mg/mL SF and 0.75, 1.5, 2.25, and 3.0 mg/mL BOA-g-HA respectively. They were lyophilized to fabricate BOA-g-HA/SF-1, BOA-g-HA/SF-2, BOA-g-HA/SF-3, and BOA-g-HA/SF-4 composite scaffolds respectively. All prepared scaffolds displayed porous network structure and exhibited promising mechanical properties for tissue engineering applications. Among them, the BOA-g-HA/SF-3 composite scaffold showed the highest influence on maintaining chondrocytic phenotype of chondrocytes under IL-1β induced stress. Following SF, HA/SF, and BOA-g-HA/SF-3 composite scaffolds with seeded chondrocytes were treated with IL-1β induction for 1 week, specimens were incubated with cell culture medium for 3 week or were subcutaneously implanted into nude mice for 4 weeks. The results demonstrated that the BOA-g-HA/SF-3 composite scaffold promotes cartilage tissue regeneration in vitro and in vivo under IL-1β caused stress, suggesting that it can be potential applied for repairing cartilage defects in osteoarthritis patients.
Collapse
Affiliation(s)
- Yongsheng Li
- College of Biology, Hunan University, Changsha 410082, PR China
| | - Xin Chen
- College of Materials Science and Engineering, Hunan University, Changsha 410082, PR China
| | - Zheng Zhou
- College of Biology, Hunan University, Changsha 410082, PR China.
| | - Bairong Fang
- Department of Plastic and Aesthetic (Burn) Surgery, the Second Xiangya Hospital, Central South University, Changsha 410001, PR China.
| | - Zongming Chen
- College of Materials Science and Engineering, Hunan University, Changsha 410082, PR China
| | - Yuting Huang
- College of Materials Science and Engineering, Hunan University, Changsha 410082, PR China
| | - Yibing Hu
- Hunan Academy of Chinese Medicine, Changsha 410013, PR China
| | - Hairong Liu
- College of Materials Science and Engineering, Hunan University, Changsha 410082, PR China
| |
Collapse
|
41
|
Feng Y, Qin S, Li H, Yang Y, Zheng Y, Liu H, Yap WY, Zhou X, Wen J. Composite Hydrogel Dressings with Enhanced Mechanical Properties and Anti-Inflammatory Ability for Effectively Promoting Wound Repair. Int J Nanomedicine 2023; 18:5183-5195. [PMID: 37720596 PMCID: PMC10503508 DOI: 10.2147/ijn.s411478] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023] Open
Abstract
Background Hydrogel dressings have been used as a crucial method to keep the wound wet and hasten the healing process. Due to safety concerns regarding the gel components, low mechanical adhesiveness, and unsatisfactory anti-inflammatory capacity qualities for practical uses in vivo, leading to the clinical translation of wound dressings is still difficult. Methods A type of composite hydrogel (acrylamide/polyethylene glycol diacrylate/tannic acid, ie, AM/PEGDA/TA) by double bond crosslinking, Schiff base, and hydrogen bond interaction is proposed. The mechanical characteristics, adhesiveness, and biocompatibility of the hydrogel system were all thoroughly examined. Additionally, a full-thickness cutaneous wound model was employed to assess the in vivo wound healing capacity of resulting hydrogel dressings. Results Benefiting the mechanism of multiple crosslinking, the designed composite hydrogels showed significant mechanical strength, outstanding adhesive capability, and good cytocompatibility. Moreover, the hydrogel system also had excellent shape adaptability, and they can be perfectly integrated into the irregularly shaped wounds through a fast in situ forming approach. Additional in vivo tests supported the findings that the full-thickness wound treated with the composite hydrogels showed quicker epithelial tissue regeneration, fewer inflammatory cells, more collagen deposition, and greater levels of platelet endothelial cell adhesion molecule (CD31) expression. Conclusion These above results might offer a practical and affordable product or method of skin wound therapy in a medical context.
Collapse
Affiliation(s)
- Yuqin Feng
- Department of Dermatology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, People’s Republic of China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, People’s Republic of China
| | - Si Qin
- Department of Dermatology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, People’s Republic of China
| | - Huarun Li
- Department of Dermatology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, People’s Republic of China
| | - Yemei Yang
- Department of Dermatology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, People’s Republic of China
| | - Yushi Zheng
- Department of Dermatology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, People’s Republic of China
| | - Hongsheng Liu
- Guangdong Huace Life Medicine Research Center, Guangzhou, 511450, People’s Republic of China
| | - Wei Yin Yap
- Department of Dermatology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, People’s Republic of China
- School of International Education of Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Xianyi Zhou
- Department of Dermatology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, People’s Republic of China
| | - Ju Wen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, People’s Republic of China
- Department of Dermatology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, People’s Republic of China
| |
Collapse
|
42
|
Leal SS, Gusmão GODM, Uchôa VT, Figueiredo-Silva J, Pinto LSS, Tim CR, Assis L, Maia-Filho ALM, de Oliveira RA, Lobo AO, Pavinatto A. Evaluation of How Methacrylate Gelatin Hydrogel Loaded with Ximenia americana L. Extract (Steam Bark) Effects Bone Repair Activity Using Rats as Models. J Funct Biomater 2023; 14:438. [PMID: 37754851 PMCID: PMC10531560 DOI: 10.3390/jfb14090438] [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: 07/05/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 09/28/2023] Open
Abstract
The use of bioactive materials, such as Ximenia americana L., to stimulate the bone repair process has already been studied; however, the synergistic effects of its association with light emitting diode (LED) have not been reported. The present work aims to evaluate the effect of its stem bark extract incorporated into methacrylate gelatin hydrogel (GelMA) on the bone repair process using pure hydrogel and hydrogel associated with LED therapy. For this purpose, the GelMA hydrogel loaded with Ximenia americana L. extract (steam bark) was produced, characterized and applied in animal experiments. The tests were performed using 50 male Wistar rats (divided into 5 groups) submitted to an induced tibia diaphyseal fracture. The therapy effects were verified for a period of 15 and 30 days of treatment using histological analysis and Raman spectroscopy. After 15 days of induced lesion/treatment, the new bone formation was significantly higher in the GXG (GelMA + X. americana L.) group compared to the control group (p < 0.0001). After 30 days, a statistically significant difference was observed when comparing the GXLEDG (GelMA + X. americana L. + LED) and the control group (p < 0.0001), the GXG and the control group (p < 0.001), and when comparing the GG, GXG (p < 0.005) and GXLEDG (p < 0.001) groups. The results shows that the Ximenia americana L. stem extract incorporated into GelMA hydrogel associated with LED therapy is a potentiator for animal bone repair.
Collapse
Affiliation(s)
- Seânia Santos Leal
- Scientific and Technological Institute, Brazil University, São Paulo 08230-030, Brazil; (S.S.L.); (C.R.T.); (L.A.)
- Biotechnology and Biodiversity Research Center, State University of Piauí, Teresina 64002-150, Brazil; (J.F.-S.); (L.S.S.P.); (A.L.M.M.-F.)
| | | | | | - José Figueiredo-Silva
- Biotechnology and Biodiversity Research Center, State University of Piauí, Teresina 64002-150, Brazil; (J.F.-S.); (L.S.S.P.); (A.L.M.M.-F.)
| | - Lucielma Salmito Soares Pinto
- Biotechnology and Biodiversity Research Center, State University of Piauí, Teresina 64002-150, Brazil; (J.F.-S.); (L.S.S.P.); (A.L.M.M.-F.)
| | - Carla R. Tim
- Scientific and Technological Institute, Brazil University, São Paulo 08230-030, Brazil; (S.S.L.); (C.R.T.); (L.A.)
| | - Lívia Assis
- Scientific and Technological Institute, Brazil University, São Paulo 08230-030, Brazil; (S.S.L.); (C.R.T.); (L.A.)
| | - Antonio Luiz Martins Maia-Filho
- Biotechnology and Biodiversity Research Center, State University of Piauí, Teresina 64002-150, Brazil; (J.F.-S.); (L.S.S.P.); (A.L.M.M.-F.)
| | | | - Anderson Oliveira Lobo
- Interdisciplinary Laboratory for Advanced Materials (LIMAV), Materials Science & Engineering Graduate Program (PPGCM), Federal University of Piauí (UFPI), Teresina 64049-550, Brazil
| | - Adriana Pavinatto
- Scientific and Technological Institute, Brazil University, São Paulo 08230-030, Brazil; (S.S.L.); (C.R.T.); (L.A.)
| |
Collapse
|
43
|
Kotla NG, Mohd Isa IL, Larrañaga A, Maddiboyina B, Swamy SK, Sivaraman G, Vemula PK. Hyaluronic Acid-Based Bioconjugate Systems, Scaffolds, and Their Therapeutic Potential. Adv Healthc Mater 2023; 12:e2203104. [PMID: 36972409 DOI: 10.1002/adhm.202203104] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/04/2023] [Indexed: 03/29/2023]
Abstract
In recent years, the development of hyaluronic acid or hyaluronan (HA) based scaffolds, medical devices, bioconjugate systems have expanded into a broad range of research and clinical applications. Research findings over the last two decades suggest that the abundance of HA in most mammalian tissues with distinctive biological roles and chemical simplicity for modifications have made it an attractive material with a rapidly growing global market. Besides its use as native forms, HA has received much interest on so-called "HA-bioconjugates" and "modified-HA systems". In this review, the importance of chemical modifications of HA, underlying rationale approaches, and various advancements of bioconjugate derivatives with their potential physicochemical, and pharmacological advantages are summarized. This review also highlights the current and emerging HA-based conjugates of small molecules, macromolecules, crosslinked systems, and surface coating strategies with their biological implications, including their potentials and key challenges discussed in detail.
Collapse
Affiliation(s)
- Niranjan G Kotla
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, 560065, India
| | - Isma Liza Mohd Isa
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, 56000, Malaysia
| | - Aitor Larrañaga
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Faculty of Engineering, University of the Basque Country (UPV/EHU), Bilbao, 48013, Spain
| | - Balaji Maddiboyina
- Department of Medical Writing, Freyr Solutions, Hyderabad, Telangana, 500081, India
| | - Samantha K Swamy
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, 9037, Norway
| | - Gandhi Sivaraman
- Department of Chemistry, Gandhigram Rural Institute (Deemed to be University), Gandhigram, Tamil Nadu, 624302, India
| | - Praveen K Vemula
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, 560065, India
| |
Collapse
|
44
|
Aldrich JL, Panicker A, Ovalle R, Sharma B. Drug Delivery Strategies and Nanozyme Technologies to Overcome Limitations for Targeting Oxidative Stress in Osteoarthritis. Pharmaceuticals (Basel) 2023; 16:1044. [PMID: 37513955 PMCID: PMC10383173 DOI: 10.3390/ph16071044] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/26/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Oxidative stress is an important, but elusive, therapeutic target for osteoarthritis (OA). Antioxidant strategies that target oxidative stress through the elimination of reactive oxygen species (ROS) have been widely evaluated for OA but are limited by the physiological characteristics of the joint. Current hallmarks in antioxidant treatment strategies include poor bioavailability, poor stability, and poor retention in the joint. For example, oral intake of exogenous antioxidants has limited access to the joint space, and intra-articular injections require frequent dosing to provide therapeutic effects. Advancements in ROS-scavenging nanomaterials, also known as nanozymes, leverage bioactive material properties to improve delivery and retention. Material properties of nanozymes can be tuned to overcome physiological barriers in the knee. However, the clinical application of these nanozymes is still limited, and studies to understand their utility in treating OA are still in their infancy. The objective of this review is to evaluate current antioxidant treatment strategies and the development of nanozymes as a potential alternative to conventional small molecules and enzymes.
Collapse
Affiliation(s)
| | | | | | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (J.L.A.)
| |
Collapse
|
45
|
Ilić-Stojanović S, Nikolić L, Cakić S. A Review of Patents and Innovative Biopolymer-Based Hydrogels. Gels 2023; 9:556. [PMID: 37504436 PMCID: PMC10378757 DOI: 10.3390/gels9070556] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
Biopolymers represent a great resource for the development and utilization of new functional materials due to their particular advantages such as biocompatibility, biodegradability and non-toxicity. "Intelligent gels" sensitive to different stimuli (temperature, pH, ionic strength) have different applications in many industries (e.g., pharmacy, biomedicine, food). This review summarizes the research efforts presented in the patent and non-patent literature. A discussion was conducted regarding biopolymer-based hydrogels such as natural proteins (i.e., fibrin, silk fibroin, collagen, keratin, gelatin) and polysaccharides (i.e., chitosan, hyaluronic acid, cellulose, carrageenan, alginate). In this analysis, the latest advances in the modification and characterization of advanced biopolymeric formulations and their state-of-the-art administration in drug delivery, wound healing, tissue engineering and regenerative medicine were addressed.
Collapse
Affiliation(s)
| | - Ljubiša Nikolić
- Faculty of Technology, University of Niš, Bulevar Oslobodjenja 124, 16000 Leskovac, Serbia
| | - Suzana Cakić
- Faculty of Technology, University of Niš, Bulevar Oslobodjenja 124, 16000 Leskovac, Serbia
| |
Collapse
|
46
|
Iaconisi GN, Lunetti P, Gallo N, Cappello AR, Fiermonte G, Dolce V, Capobianco L. Hyaluronic Acid: A Powerful Biomolecule with Wide-Ranging Applications-A Comprehensive Review. Int J Mol Sci 2023; 24:10296. [PMID: 37373443 DOI: 10.3390/ijms241210296] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Hyaluronic acid (HA) is a glycosaminoglycan widely distributed in the human body, especially in body fluids and the extracellular matrix of tissues. It plays a crucial role not only in maintaining tissue hydration but also in cellular processes such as proliferation, differentiation, and the inflammatory response. HA has demonstrated its efficacy as a powerful bioactive molecule not only for skin antiaging but also in atherosclerosis, cancer, and other pathological conditions. Due to its biocompatibility, biodegradability, non-toxicity, and non-immunogenicity, several HA-based biomedical products have been developed. There is an increasing focus on optimizing HA production processes to achieve high-quality, efficient, and cost-effective products. This review discusses HA's structure, properties, and production through microbial fermentation. Furthermore, it highlights the bioactive applications of HA in emerging sectors of biomedicine.
Collapse
Affiliation(s)
- Giorgia Natalia Iaconisi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | - Paola Lunetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | - Nunzia Gallo
- Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy
| | - Anna Rita Cappello
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Giuseppe Fiermonte
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Vincenza Dolce
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Loredana Capobianco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| |
Collapse
|
47
|
Aunina K, Ramata-Stunda A, Kovrlija I, Tracuma E, Merijs-Meri R, Nikolajeva V, Loca D. Exploring the Interplay of Antimicrobial Properties and Cellular Response in Physically Crosslinked Hyaluronic Acid/ε-Polylysine Hydrogels. Polymers (Basel) 2023; 15:polym15081915. [PMID: 37112064 PMCID: PMC10141856 DOI: 10.3390/polym15081915] [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: 02/27/2023] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
The reduction of tissue cytotoxicity and the improvement of cell viability are of utmost significance, particularly in the realm of green chemistry. Despite substantial progress, the threat of local infections remains a concern. Therefore, hydrogel systems that provide mechanical support and a harmonious balance between antimicrobial efficacy and cell viability are greatly needed. Our study explores the preparation of physically crosslinked, injectable, and antimicrobial hydrogels using biocompatible hyaluronic acid (HA) and antimicrobial ε-polylysine (ε-PL) in different weight ratios (10 wt% to 90 wt%). The crosslinking was achieved by forming a polyelectrolyte complex between HA and ε-PL. The influence of HA content on the resulting HA/ε-PL hydrogel physicochemical, mechanical, morphological, rheological, and antimicrobial properties was evaluated, followed by an inspection of their in vitro cytotoxicity and hemocompatibility. Within the study, injectable, self-healing HA/ε-PL hydrogels were developed. All hydrogels showed antimicrobial properties against S. aureus, P. aeruginosa, E. coli, and C. albicans, where HA/ε-PL 30:70 (wt%) composition reached nearly 100% killing efficiency. The antimicrobial activity was directly proportional to ε-PL content in the HA/ε-PL hydrogels. A decrease in ε-PL content led to a reduction of antimicrobial efficacy against S. aureus and C. albicans. Conversely, this decrease in ε-PL content in HA/ε-PL hydrogels was favourable for Balb/c 3T3 cells, leading to the cell viability of 152.57% for HA/ε-PL 70:30 and 142.67% for HA/ε-PL 80:20. The obtained results provide essential insights into the composition of the appropriate hydrogel systems able to provide not only mechanical support but also the antibacterial effect, which can offer opportunities for developing new, patient-safe, and environmentally friendly biomaterials.
Collapse
Affiliation(s)
- Kristine Aunina
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, LV-1007 Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, LV-1007 Riga, Latvia
| | - Anna Ramata-Stunda
- Department of Microbiology and Biotechnology, Faculty of Biology, University of Latvia, LV-1050 Riga, Latvia
| | - Ilijana Kovrlija
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, LV-1007 Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, LV-1007 Riga, Latvia
| | - Eliza Tracuma
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, LV-1007 Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, LV-1007 Riga, Latvia
| | - Remo Merijs-Meri
- Institute of Polymer Materials, Faculty of Materials Science and Applied Chemistry, Riga Technical University, LV-1048 Riga, Latvia
| | - Vizma Nikolajeva
- Department of Microbiology and Biotechnology, Faculty of Biology, University of Latvia, LV-1050 Riga, Latvia
| | - Dagnija Loca
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, LV-1007 Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, LV-1007 Riga, Latvia
| |
Collapse
|
48
|
Jahanbekam S, Mozafari N, Bagheri-Alamooti A, Mohammadi-Samani S, Daneshamooz S, Heidari R, Azarpira N, Ashrafi H, Azadi A. Ultrasound-responsive hyaluronic acid hydrogel of hydrocortisone to treat osteoarthritis. Int J Biol Macromol 2023; 240:124449. [PMID: 37072059 DOI: 10.1016/j.ijbiomac.2023.124449] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/20/2023]
Abstract
One of the practical ways to manage the disease flares of arthritis is using an intra-articular depot formulation of glucocorticoids. Hydrogels, as controllable drug delivery systems, are hydrophilic polymers with distinctive properties, such as remarkable water capacity and biocompatibility. This study aimed to design an injectable thermo-ultrasound-triggered drug carrier based on Pluronic® F-127, hyaluronic acid, and gelatin. The in situ hydrogel loaded by hydrocortison was developed and D-optimal design was used to formulate the process. The optimized hydrogel was combined with four different surfactants to better regulate the release rate. In situ gels composed of the hydrocortisone-loaded hydrogel and hydrocortisone-loaded mixed-micelle hydrogel were characterized. The hydrocortisone-loaded hydrogel and selected hydrocortisone-loaded mixed-micelle hydrogel showed a spherical shape and were nano-sized with a unique thermo-responsive nature able to prolong drug release. The ultrasound-triggered release study showed that drug release was time-dependent. By inducing osteoarthritis in a rat model, behavioral tests and histopathological analyses were carried out on the hydrocortisone-loaded hydrogel and a particular hydrocortisone-loaded mixed-micelle hydrogel. In vivo results showed that the selected hydrocortisone-loaded mixed-micelle hydrogel improved the status of the disease. Results highlighted the potential of ultrasound-responsive in situ-forming hydrogels as hopeful formulas for efficient treatment of arthritis.
Collapse
Affiliation(s)
- Sheida Jahanbekam
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Mozafari
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azar Bagheri-Alamooti
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soliman Mohammadi-Samani
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Daneshamooz
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hajar Ashrafi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Azadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
49
|
Lee UJ, Sohng JK, Kim BG, Choi KY. Recent trends in the modification of polyphenolic compounds using hydroxylation and glycosylation. Curr Opin Biotechnol 2023; 80:102914. [PMID: 36857963 DOI: 10.1016/j.copbio.2023.102914] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/08/2023] [Accepted: 01/31/2023] [Indexed: 03/02/2023]
Abstract
Polyphenols are bioactive molecules that are used in therapeutics. Polyphenol hydroxylation and glycosylation have been shown to increase their bioavailability, solubility, bioactivity, and stability for use in various applications. Ortho-hydroxylation of polyphenols using tyrosinase allows high selectivity and yield without requiring a cofactor, while meta- and para-hydroxylation of polyphenols are mediated by site-specific hydroxylases and cytochrome P450s, although these processes are somewhat rare. O-glycosylation of polyphenols proceeds further after hydroxylation. The O-glycosylation reaction typically requires nucleotide diphosphate (NDP) sugar. However, amylosucrase (AS) has emerged as a promising enzyme for polyphenol glycosylation in large-scale production without requiring NDP-sugar. Overall, this review describes recent findings on the enzymatic mechanisms, enzyme engineering, and applications of enzymatic reactions.
Collapse
Affiliation(s)
- Uk-Jae Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX/N-Bio, Institute of BioEngineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Kyung Sohng
- Institute of Biomolecule Reconstruction (iBR), Department of Life Science and Biochemical Engineering, Sun Moon University, Asan-si, Chungnam, Republic of Korea; Department of Biotechnology and Pharmaceutical Engineering, Sun Moon University, Asan-si, Chungnam, Republic of Korea
| | - Byung-Gee Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX/N-Bio, Institute of BioEngineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Kwon-Young Choi
- Department of Environmental and Safety Engineering, College of Engineering, Ajou University, Republic of Korea; Department of Energy Systems Research, Ajou University, Republic of Korea.
| |
Collapse
|
50
|
Deng H, Wang J, An R. Hyaluronic acid-based hydrogels: As an exosome delivery system in bone regeneration. Front Pharmacol 2023; 14:1131001. [PMID: 37007032 PMCID: PMC10063825 DOI: 10.3389/fphar.2023.1131001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/03/2023] [Indexed: 03/19/2023] Open
Abstract
Exosomes are extracellular vesicles (EVs) containing various ingredients such as DNA, RNA, lipids and proteins, which play a significant role in intercellular communication. Numerous studies have demonstrated the important role of exosomes in bone regeneration through promoting the expression of osteogenic-related genes and proteins in mesenchymal stem cells. However, the low targeting ability and short circulating half-life of exosomes limited their clinical application. In order to solve those problems, different delivery systems and biological scaffolds have been developed. Hydrogel is a kind of absorbable biological scaffold composed of three-dimensional hydrophilic polymers. It not only has excellent biocompatibility and superior mechanical strength but can also provide a suitable nutrient environment for the growth of the endogenous cells. Thus, the combination between exosomes and hydrogels can improve the stability and maintain the biological activity of exosomes while achieving the sustained release of exosomes in the bone defect sites. As an important component of the extracellular matrix (ECM), hyaluronic acid (HA) plays a critical role in various physiological and pathological processes such as cell differentiation, proliferation, migration, inflammation, angiogenesis, tissue regeneration, wound healing and cancer. In recent years, hyaluronic acid-based hydrogels have been used as an exosome delivery system for bone regeneration and have displayed positive effects. This review mainly summarized the potential mechanism of HA and exosomes in promoting bone regeneration and the application prospects and challenges of hyaluronic acid-based hydrogels as exosome delivery devices in bone regeneration.
Collapse
Affiliation(s)
| | | | - Ran An
- *Correspondence: Jiecong Wang, ; Ran An,
| |
Collapse
|