1
|
Guan Q, Hou S, Wang K, Li L, Cheng Y, Zheng M, Liu C, Zhao X, Zhou J, Li P, Niu X, Wang L, Fan Y. Micropore structure engineering of injectable granular hydrogels via controlled liquid-liquid phase separation facilitates regenerative wound healing in mice and pigs. Biomaterials 2025; 318:123192. [PMID: 39965423 DOI: 10.1016/j.biomaterials.2025.123192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/22/2025] [Accepted: 02/13/2025] [Indexed: 02/20/2025]
Abstract
Biomaterials can play a crucial role in facilitating tissue regeneration, but their application is often limited by that they induce scarring rather than complete tissue restoration. Hydrogels with microporous architectures, engineered via 3D printing techniques or particle packing (granular hydrogels), have shown promise in providing a conducive microenvironment for cellular infiltration and favorable immune response. Nonetheless, there is a notably lacking in studies that demonstrate scarless regeneration solely through pore structure engineering. In this study, we demonstrate that optimizing micropore structure of injectable granular hydrogels via controlled liquid-liquid phase separation facilitates scarless wound healing. The building block particles are fabricated by precisely controlling the separation kinetics of two immiscible aqueous phases (gelling and porogenic) and timely arresting phase separation, to generate bicontinuous, hollow or closed porous structure. Employing a murine model, we reveal that the optimized pore structure significantly facilitates mature vascular network boosts pro-regenerative macrophage polarization (M2/M1) and CD4+/Foxp3+ regulatory T cells, culminating in scarless skin regeneration enriched with hair follicles. Moreover, our hydrogels outperform the clinical gold-standard collagen/proteoglycan scaffolds in a porcine model, showcasing superior cell infiltration, epidermal integration, and dermal regeneration. Micropore structure engineering of biomaterials presents a promising and biologics free pathway for tissue regeneration.
Collapse
Affiliation(s)
- Qifeng Guan
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115, Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Sen Hou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China.
| | - Kai Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Linhao Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yating Cheng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Mingxia Zheng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Chen Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xinbin Zhao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Jin Zhou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Ping Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xufeng Niu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Lizhen Wang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115, Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yubo Fan
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, 311115, Hangzhou, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological and Medical Engineering, Beihang University, Beijing, 100191, China.
| |
Collapse
|
2
|
Wang H, Cheng P, Wang J, Lv H, Han J, Hou Z, Xu R, Chen W. Advances in spatial transcriptomics and its application in the musculoskeletal system. Bone Res 2025; 13:54. [PMID: 40379648 DOI: 10.1038/s41413-025-00429-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 05/19/2025] Open
Abstract
While bulk RNA sequencing and single-cell RNA sequencing have shed light on cellular heterogeneity and potential molecular mechanisms in the musculoskeletal system in both physiological and various pathological states, the spatial localization of cells and molecules and intercellular interactions within the tissue context require further elucidation. Spatial transcriptomics has revolutionized biological research by simultaneously capturing gene expression profiles and in situ spatial information of tissues, gradually finding applications in musculoskeletal research. This review provides a summary of recent advances in spatial transcriptomics and its application to the musculoskeletal system. The classification and characteristics of data acquisition techniques in spatial transcriptomics are briefly outlined, with an emphasis on widely-adopted representative technologies and the latest technological breakthroughs, accompanied by a concise workflow for incorporating spatial transcriptomics into musculoskeletal system research. The role of spatial transcriptomics in revealing physiological mechanisms of the musculoskeletal system, particularly during developmental processes, is thoroughly summarized. Furthermore, recent discoveries and achievements of this emerging omics tool in addressing inflammatory, traumatic, degenerative, and tumorous diseases of the musculoskeletal system are compiled. Finally, challenges and potential future directions for spatial transcriptomics, both as a field and in its applications in the musculoskeletal system, are discussed.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Peng Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Juan Wang
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Hongzhi Lv
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Jie Han
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhiyong Hou
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| | - Wei Chen
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China.
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China.
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China.
| |
Collapse
|
3
|
Xu C, Wu F, Duan Z, Rajbanshi B, Qi Y, Qin J, Dai L, Liu C, Jin T, Zhang B, Zhang X. Microneedle-aided nanotherapeutics delivery and nanosensor intervention in advanced tissue regeneration. J Nanobiotechnology 2025; 23:330. [PMID: 40319333 PMCID: PMC12048949 DOI: 10.1186/s12951-025-03383-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/10/2025] [Indexed: 05/07/2025] Open
Abstract
Microneedles (MNs) have been extensively used as transdermal therapeutics delivery devices since 1998 due to their capacity to penetrate physiological barriers with minimal invasiveness. Recent advances demonstrate the potential of MNs in improving diverse tissue regeneration when integrated with nanometer-sized therapeutics or sensors. This synergistic strategy can enhance drug delivery efficiency and therapeutic outcomes, and enable precise and personalized therapies through real-time monitoring of the repair process. In this review, we discuss how optimized MNs (through adjustments in geometry, material properties, and modular structure), when combined with dimension- and composition-specific nanomaterials, enhance tissue regeneration efficiency. Moreover, integrating stimuli-responsive nanotherapeutics or nanosensors into MNs for spatiotemporal-controlled and targeted drug release, physiotherapy effects, and intelligent monitoring is systematically outlined. Furthermore, we summarize therapeutic applications of nanotherapeutics-MN platforms in various soft and hard tissues, including skin, hair follicles (HF), cornea, joint, tendons, sciatic nerves, spinal cord, periodontium, oral mucosa, myocardium, endometrium, bone and intervertebral discs (IVD). Notably, recent attempts using nanosensor-MN platforms as smart wearable devices for monitoring damaged tissues via interstitial fluid (ISF) extraction and biomarker sensing are analyzed. This review potentially provides tissue regeneration practitioners/researchers with a cross-disciplinary perspective and inspiration.
Collapse
Affiliation(s)
- Churong Xu
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Fei Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhouyi Duan
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Bhavana Rajbanshi
- Department of Dermatology and Venereology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yuxin Qi
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jiaming Qin
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Liming Dai
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Chaozong Liu
- Institute of Orthopaedic & Musculoskeletal Science, University College London, Royal National Orthopaedic Hospital, Stanmore, HA7 4LP, UK
| | - Tuo Jin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Bingjun Zhang
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Xiaoling Zhang
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
4
|
Xu Z, Yang Y, Li X, Wang J, Chen S, An T, Hu C, Deng C, Zhou F, Xiang L, Qu Y, Man Y. A Visible-Light Photocatalysis/Hydrolysis Hydrogen-Generating Nanoplatform for Dynamic Inflammation Management via Immune Metabolism Orchestration during Wound Repair. ACS APPLIED MATERIALS & INTERFACES 2025; 17:24918-24939. [PMID: 40117501 DOI: 10.1021/acsami.5c00667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Effective management of inflammation is one of the promising strategies to prevent the formation of chronic wounds. Despite hydrogen being a prospective molecule for anti-inflammatory effects, the on-demand delivery of hydrogen that could synchronize with the dynamic inflammation stages has yet remained unaddressed. Moreover, its specific immunomodulatory mechanisms are still veiled. In this study, we introduced ISO-ZIF-8@AB, a hydrogen-generating nanoplatform that integrated visible-light photocatalysis and hydrolysis reactions to achieve controllable hydrogen release on demand, functioning with an initial peak release and following a sustained release. With ISO-ZIF-8@AB further loaded into an aligned ECM-like scaffold, the complex significantly alleviated inflammation and prevented protracted unhealing. The bulk-RNA sequencing combined with single-cell RNA sequencing revealed that hydrogen treatment effectively reduced the excessive aggregation and infiltration of innate immune cells. Specifically, hydrogen reduced the proportion of Ptgs2+Nos2+ pro-inflammatory macrophages (PIMs) by mitigating mitochondrial stress and suppressing HIF-1α-induced glycolysis, the immune-metabolic regulation of which reduced harmful crosstalk between PIMs and hypodermal fibroblasts and facilitated extracellular matrix production accompanied by the ultimate wound repair. Overall, this study presented a strategy for controllable hydrogen release in terms of timing and rate, with further discussions regarding the underlying immune-metabolic regulation mechanisms of hydrogen therapy.
Collapse
Affiliation(s)
- Zhaoyu Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yang Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinhui Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuaidong Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tiantian An
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chen Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chen Deng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Feng Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yili Qu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yi Man
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
5
|
Vasan A, Kim S, Davis E, Roh DS, Eyckmans J. Advances in Designer Materials for Chronic Wound Healing. Adv Wound Care (New Rochelle) 2025. [PMID: 40306934 DOI: 10.1089/wound.2024.0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Significance: Nonhealing or chronic wounds represent a significant and growing global health concern, imposing substantial burdens on individuals, health care systems, and economies worldwide. Although the standard-of-care treatment involves the application of wound dressings, most dressing materials are not specifically designed to address the pathological processes underlying chronic wounds. This review highlights recent advances in biomaterial design tailored to chronic wound healing. Recent Advances: Chronic wounds are characterized by persistent inflammation, impaired granulation tissue formation, and delayed re-epithelialization. Newly developed designer materials aim to manage reactive oxygen species and extracellular matrix degradation to suppress inflammation while promoting vascularization, cell proliferation, and epithelial migration to accelerate tissue repair. Critical Issues: Designing optimal materials for chronic wounds remains challenging due to the diverse etiology and a multitude of pathological mechanisms underlying chronic wound healing. While designer materials can target specific aberrations, designing a materials approach that restores all aberrant wound-healing processes remains the Holy Grail. Addressing these issues requires a deep understanding of how cells interact with the materials and the complex etiology of chronic wounds. Future Directions: New material approaches that target wound mechanics and senescence to improve chronic wound closure are under development. Layered materials combining the best properties of the approaches discussed in this review will pave the way for designer materials optimized for chronic wound healing.
Collapse
Affiliation(s)
- Anish Vasan
- Department of Biomedical Engineering and the Biological Design Center, Boston University, Boston, Massachusetts, USA
| | - Suntae Kim
- Department of Biomedical Engineering and the Biological Design Center, Boston University, Boston, Massachusetts, USA
| | - Emily Davis
- Department of Biomedical Engineering and the Biological Design Center, Boston University, Boston, Massachusetts, USA
| | - Daniel S Roh
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jeroen Eyckmans
- Department of Biomedical Engineering and the Biological Design Center, Boston University, Boston, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Zhang C, Song W, Guo X, Li Z, Kong Y, Du J, Hou L, Feng Y, Wang Y, Zhang M, Liang L, Huang Y, Li J, Zhu D, Liu Q, Tan Y, Zhao Z, Zhao Y, Fu X, Huang S. Piezoelectric nanocomposite electrospun dressings: Tailoring mechanics for scar-free wound recovery. BIOMATERIALS ADVANCES 2025; 167:214119. [PMID: 39556886 DOI: 10.1016/j.bioadv.2024.214119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
Rational wound management and enhancing healing quality are critical in clinical practice. Electrical stimulation therapy (EST) has emerged as a valuable adjunctive treatment due to its safety and cost-effectiveness. Integrating piezoelectric materials into dressings offers a way to miniaturize and personalize electrotherapy, enhancing convenience. To address the impact of physical factors of dressings on wound healing, a nanocomposite piezoelectric electrospun dressing using poly(L-lactic acid) (PLLA) and barium titanate (BaTiO3) was developed. We intentionally exaggerated design flaws to mimic the characteristics of scar extracellular matrix (ECM), including the oriented thick fibers and high Young's modulus. Initially, these dressings promoted fibrosis and hindered functional regeneration. However, when the piezoelectric effect was triggered by ultrasound, the fibrotic phenotype was reversed, leading to scar-free healing with well-regenerated functional structures. This study highlights the significant therapeutic potential of piezoelectric dressings in skin wound treatment and underscores the importance of carefully designing the static physical properties of dressings for optimal efficacy.
Collapse
Affiliation(s)
- Chao Zhang
- School of Medicine, Nankai University, Tianjin 300071, China; Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Wei Song
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Xu Guo
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China; College of Graduate, Tianjin Medical University, Tianjin 300203, China
| | - Zhao Li
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Yi Kong
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Jinpeng Du
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Linhao Hou
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Yu Feng
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Yuzhen Wang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Mengde Zhang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Liting Liang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Yuyan Huang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Jianjun Li
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Dongzhen Zhu
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Qinghua Liu
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Yaxin Tan
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China
| | - Ziteng Zhao
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yantao Zhao
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Xiaobing Fu
- School of Medicine, Nankai University, Tianjin 300071, China; Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China; College of Graduate, Tianjin Medical University, Tianjin 300203, China.
| | - Sha Huang
- Research Center for Wound Repair and Tissue Regeneration, Medical Innovation Research Department, Chinese PLA General Hospital, Beijing 100048, China; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing 100048, China.
| |
Collapse
|
7
|
Luo W, Ren L, Hu B, Zhang H, Yang Z, Jin L, Zhang D. Recent Development of Fibrous Hydrogels: Properties, Applications and Perspectives. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408657. [PMID: 39530645 PMCID: PMC11714238 DOI: 10.1002/advs.202408657] [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: 07/26/2024] [Revised: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Fibrous hydrogels (FGs), characterized by a 3D network structure made from prefabricated fibers, fibrils and polymeric materials, have emerged as significant materials in numerous fields. However, the challenge of balancing mechanical properties and functions hinders their further development. This article reviews the main advantages of FGs, including enhanced mechanical properties, high conductivity, high antimicrobial and anti-inflammatory properties, stimulus responsiveness, and an extracellular matrix (ECM)-like structure. It also discusses the influence of assembly methods, such as fiber cross-linking, interfacial treatments of fibers with hydrogel matrices, and supramolecular assembly, on the diverse functionalities of FGs. Furthermore, the mechanisms for improving the performance of the above five aspects are discussed, such as creating ion carrier channels for conductivity, in situ gelation of drugs to enhance antibacterial and anti-inflammatory properties, and entanglement and hydrophobic interactions between fibers, resulting in ECM-like structured FGs. In addition, this review addresses the application of FGs in sensors, dressings, and tissue scaffolds based on the synergistic effects of optimizing the performance. Finally, challenges and future applications of FGs are discussed, providing a theoretical foundation and new insights for the design and application of cutting-edge FGs.
Collapse
Affiliation(s)
- Wen Luo
- International Joint Research Laboratory for Biomedical Nanomaterials of HenanHenan Key Laboratory of Rare Earth Functional MaterialsZhoukou Normal UniversityZhoukou466001P. R. China
| | - Liujiao Ren
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Bin Hu
- International Joint Research Laboratory for Biomedical Nanomaterials of HenanHenan Key Laboratory of Rare Earth Functional MaterialsZhoukou Normal UniversityZhoukou466001P. R. China
| | - Huali Zhang
- International Joint Research Laboratory for Biomedical Nanomaterials of HenanHenan Key Laboratory of Rare Earth Functional MaterialsZhoukou Normal UniversityZhoukou466001P. R. China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Research Institute of Xi'an Jiaotong UniversityHangzhou311200P. R. China
| | - Lin Jin
- International Joint Research Laboratory for Biomedical Nanomaterials of HenanHenan Key Laboratory of Rare Earth Functional MaterialsZhoukou Normal UniversityZhoukou466001P. R. China
| | - Di Zhang
- Department of General Surgery (Colorectal Surgery)Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesGuangdong Institute of GastroenterologyBiomedical Innovation Center, The Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhou510655P. R. China
| |
Collapse
|
8
|
Dai X, Nie W, Shen H, Machens HG, Böker K, Taheri S, Lehmann W, Shen Y, Schilling AF. Electrospinning based biomaterials for biomimetic fabrication, bioactive protein delivery and wound regenerative repair. Regen Biomater 2024; 12:rbae139. [PMID: 39803356 PMCID: PMC11723536 DOI: 10.1093/rb/rbae139] [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: 07/28/2024] [Revised: 10/25/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Electrospinning is a remarkably straightforward and adaptable technique that can be employed to process an array of synthetic and natural materials, resulting in the production of nanoscale fibers. It has emerged as a novel technique for biomedical applications and has gained increasing popularity in the research community in recent times. In the context of tissue repair and tissue engineering, there is a growing tendency toward the integration of biomimetic scaffolds and bioactive macromolecules, particularly proteins and growth factors. The design of 'smart' systems provides not merely physical support, but also microenvironmental cues that can guide regenerative tissue repair. Electrospun nanofibrous matrices are regarded as a highly promising tool in this area, as they can serve as both an extracellular matrix (ECM)-mimicking scaffold and a vehicle for the delivery of bioactive proteins. Their highly porous architecture and high surface-to-volume ratio facilitate the loading of drugs and mass transfer. By employing a judicious selection of materials and processing techniques, there is considerable flexibility in efficiently customizing nanofiber architecture and incorporating bioactive proteins. This article presents a review of the strategies employed for the structural modification and protein delivery of electrospun nanofibrous materials, with a focus on the objective of achieving a tailored tissue response. The article goes on to discuss the challenges currently facing the field and to suggest future research directions.
Collapse
Affiliation(s)
- Xinyi Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wei Nie
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, NC 27103, USA
| | - Hua Shen
- Department of Plastic and Reconstructive Surgery, Shanghai First People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, Faculty of Medicine, Technical University of Munich, Munich 81675, Germany
| | - Kai Böker
- Department of Trauma Surgery, Orthopaedic Surgery and Plastic Surgery, University Medical Center Göttingen, University of Göttingen, Göttingen 37075, Germany
| | - Shahed Taheri
- Department of Trauma Surgery, Orthopaedic Surgery and Plastic Surgery, University Medical Center Göttingen, University of Göttingen, Göttingen 37075, Germany
| | - Wolfgang Lehmann
- Department of Trauma Surgery, Orthopaedic Surgery and Plastic Surgery, University Medical Center Göttingen, University of Göttingen, Göttingen 37075, Germany
| | - Yi Shen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Arndt F Schilling
- Department of Trauma Surgery, Orthopaedic Surgery and Plastic Surgery, University Medical Center Göttingen, University of Göttingen, Göttingen 37075, Germany
| |
Collapse
|
9
|
Zivari-Ghader T, Shokouhi B, Kosari-Nasab M, Davaran S, Hamishehkar H, Farahpour MR, Rashidi MR, Mehrali M. Hypericum Perforatum Callus Extract-Loaded Composite Hydrogel with Diverse Bioactivities for Enhanced Wound Healing and Fibrosis Prevention. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2407112. [PMID: 39498666 DOI: 10.1002/smll.202407112] [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: 08/15/2024] [Revised: 10/07/2024] [Indexed: 11/07/2024]
Abstract
Plant Callus are a valuable source of pluripotent stem cells and bioactive phytochemicals. Meanwhile, the Hypericum perforatum callus extract (HPCE) is particularly rich in compounds such as hyperforin, hypericin, quercetin, and other phenolic and flavonoid derivatives. These phytochemicals exhibit strong antibacterial, antioxidant, anti-inflammatory, and anti-fibrotic properties, making them promising for wound healing. One of the most critical challenges following wound healing is the formation of fibrosis, which can compromise the complex structural integrity of skin. To address this issue, a poly(vinyl alcohol)/chitosan/alginate (PCA) wound dressing loaded with HPCE is developed. This hydrogel dressing features a porous structure with suitable mechanical properties and a high swelling capacity, potentially enhancing its effectiveness in promoting tissue regeneration and wound healing. In vitro studies have confirmed its biocompatibility, cell proliferation, and cell adhesion properties. Additionally, the dressing has demonstrated the ability to inhibit the proliferation of certain antibiotic-resistant bacteria. The in vivo studies revealed the anti-inflammatory properties, promotion of angiogenesis, facilitation of re-epithelialization, and stimulation of collagen deposition of the dressing under investigation. Moreover, the immunohistochemistry analysis of the two key markers, p16 and p53, has shown that the application of the dressing helps prevent fibrosis after wound healing.
Collapse
Affiliation(s)
- Tayebeh Zivari-Ghader
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, 51664-14766, Iran
| | - Behrooz Shokouhi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, 51664-14766, Iran
| | - Morteza Kosari-Nasab
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, 51666-16471, Iran
| | - Soodabeh Davaran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, 51664-14766, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, 51656-65811, Iran
| | - Mohammad Reza Farahpour
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, 57159-14338, Iran
| | - Mohammad-Reza Rashidi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, 51664-14766, Iran
| | - Mehdi Mehrali
- Department of Civil and Mechanical Engineering, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| |
Collapse
|
10
|
Tao Q, Liang Q, Fu Y, Qian J, Xu J, Zhu Y, Gu C, Xu W, Song S, Wu Y, Wang Y, Peng Y, Wang L, Gao Q. Puerarin ameliorates colitis by direct suppression of macrophage M1 polarization in DSS mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156048. [PMID: 39326132 DOI: 10.1016/j.phymed.2024.156048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a type of inflammatory bowel disease primarily affecting the colon and rectum. The clinical symptoms of UC include persistent diarrhea, abdominal pain, and rectal bleeding, with chronic inflammation often limited to the mucosal layer of the colon. Macrophages play a significant role in the pathogenesis of UC in response to the presence of gut microbiota. Puerarin is an active compound derived from the root of pueraria lobata, a traditional Chinese herbal medicine, and exhibits potent anti-inflammatory properties in various diseases and disease models including UC-like colitis in mice. However, how the molecule achieves its therapeutic effect in colitis by re-polarizing macrophages remains poorly understood. PURPOSE Utilizing in vivo and in vitro experimental methods along with multi-omics analysis, we aimed to elucidate the potential mechanism by which puerarin targets macrophages to treat colitis. METHODS The inflammation induced by DSS was assessed both locally in the gut and systemically, and the anti-inflammatory effect of puerarin was evaluated using molecular and histological assays such as H&E staining, qPCR, ELISA, Western blot, and immunofluorescence. Intestinal permeability parameters were measured by in vivo imaging, immunofluorescence, Western blot, qPCR, and PAS staining. The central role of macrophages in colitis was investigated through macrophage depletion/infusion using cytological methods. The direct effects of puerarin on the macrophages were examined by CCK8, flow cytometry, and qPCR in vitro. Additionally, 16S rRNA sequencing and metabolomics analysis of gut contents were conducted. Identification of key pathogenic flora was facilitated by a trans-omic approach and validated both in vitro and in vivo. RESULTS Puerarin exerted a direct and robust suppression of M1-like polarization of macrophages in vitro, which was sufficient to confer therapeutic benefits in terms of colonic lesions and systemic inflammation in DSS mice. Puerarin also reduced the abundance of Akkermansia muciniphila in the gut, which was significantly upregulated in DSS mice in our experimental context. Further study demonstrated that puerarin effectively suppressed M1-like macrophage activation induced by Akkermansia muciniphila secreted protein Amuc_2172, thereby altering the pathology in the DSS model. CONCLUSION Our data suggest that the pathogenesis of DSS colitis is mediated by host cellular responses to toxic foreign molecules and the gut microbiota, and targeting specific cell populations, such as macrophages, with puerarin holds potential therapeutic value.
Collapse
Affiliation(s)
- Qing Tao
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Qiao Liang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yu Fu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Jun Qian
- Nangjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu 210008, China
| | - Jing Xu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yimin Zhu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Chinese Medicine and Jiangsu Second Chinese Medicine Hospital, 23 Hunan Road, Nanjing 210017, China
| | - Cheng Gu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Chinese Medicine and Jiangsu Second Chinese Medicine Hospital, 23 Hunan Road, Nanjing 210017, China
| | - Wenhui Xu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Chinese Medicine and Jiangsu Second Chinese Medicine Hospital, 23 Hunan Road, Nanjing 210017, China
| | - Shiyu Song
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yongzheng Wu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yong Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yuming Peng
- Department of Cadre Health Care, Karamay Central Hospital, No. 67, Junggar Road, Xinjiang 834000, China
| | - Lei Wang
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China.
| |
Collapse
|
11
|
Chen SD, Chu CY, Wang CB, Yang Y, Xu ZY, Qu YL, Man Y. Integrated-omics profiling unveils the disparities of host defense to ECM scaffolds during wound healing in aged individuals. Biomaterials 2024; 311:122685. [PMID: 38944969 DOI: 10.1016/j.biomaterials.2024.122685] [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: 02/05/2024] [Revised: 06/11/2024] [Accepted: 06/23/2024] [Indexed: 07/02/2024]
Abstract
Extracellular matrix (ECM) scaffold membranes have exhibited promising potential to better the outcomes of wound healing by creating a regenerative microenvironment around. However, when compared to the application in younger individuals, the performance of the same scaffold membrane in promoting re-epithelialization and collagen deposition was observed dissatisfying in aged mice. To comprehensively explore the mechanisms underlying this age-related disparity, we conducted the integrated analysis, combing single-cell RNA sequencing (scRNA-Seq) with spatial transcriptomics, and elucidated six functionally and spatially distinctive macrophage groups and lymphocytes surrounding the ECM scaffolds. Through intergroup comparative analysis and cell-cell communication, we characterized the dysfunction of Spp1+ macrophages in aged mice impeded the activation of the type Ⅱ immune response, thus inhibiting the repair ability of epidermal cells and fibroblasts around the ECM scaffolds. These findings contribute to a deeper understanding of biomaterial applications in varied physiological contexts, thereby paving the way for the development of precision-based biomaterials tailored specifically for aged individuals in future therapeutic strategies.
Collapse
Affiliation(s)
- Shuai-Dong Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen-Yu Chu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen-Bing Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Yang Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhao-Yu Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi-Li Qu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Man
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
12
|
Zeng K, Lin Y, Liu S, Wang Z, Guo L. Applications of piezoelectric biomaterials in dental treatments: A review of recent advancements and future prospects. Mater Today Bio 2024; 29:101288. [PMID: 40018432 PMCID: PMC11866170 DOI: 10.1016/j.mtbio.2024.101288] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 03/01/2025] Open
Abstract
Piezoelectric biomaterials have attracted considerable attention in dental medicine due to their unique ability to convert mechanical force into electricity and catalyze reactions. These materials demonstrate biocompatibility, high bioactivity, and stability, making them suitable for applications such as tissue regeneration, caries prevention, and periodontal disease treatment. Despite their significant potential, the clinical application of these materials in treating oral diseases remains limited, facing numerous challenges in clinical translation. Therefore, further research and data are crucial to advance their application in dentistry. The review emphasizes the transformative impact of multifunctional piezoelectric biomaterials on enhancing dental therapies and outlines future directions for their integration into oral healthcare practices.
Collapse
Affiliation(s)
- Kaichen Zeng
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yifan Lin
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shirong Liu
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ziyan Wang
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lvhua Guo
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Li X, Zhang KW, Zhang ZY, Wu JJ, Yuan ZD, Yuan FL, Chen J. Inhibiting dipeptidyl peptidase 4 positive fibroblasts using zinc sulfide cellulose nanofiber scaffolds to achieve scarless healing. Int J Biol Macromol 2024; 282:137525. [PMID: 39537047 DOI: 10.1016/j.ijbiomac.2024.137525] [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: 08/14/2024] [Revised: 11/05/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Wound regeneration with integral function and cutaneous appendages remains challenging in wound dressing applications. Cellulose nanofibers (CNF) exhibit remarkable characteristics in wound dressing applications; however, their utility in the wound healing process is limited by insufficient scar inhibition and regenerative healing. Herein, inspired by fibroblast heterogeneity mediating wound healing and skin regeneration, we developed a CNF scaffold designed to block Dipeptidyl Peptidase 4 positive (DPP4+) fibroblasts for regenerative healing. CNF encapsulated sitagliptin (SITA) and zinc sulfide nanoparticles (NZnS), namely CNF@SITA@NZnS, to fabricate a novel biomaterial for scar reduction and regenerative healing. The scaffold promoted scarless healing and hair follicle regeneration in rats. In vivo experiments, the wounds in the scaffold showed less skin fibrosis, a better collagen ratio and more new hair follicles. In vitro experiments showed that the scaffold material promoted scarless healing, possibly by inhibiting the secretion of extracellular matrix and fibroblast-to-myofibroblast conversion. The promotion of hair follicle regeneration by the scaffold material may be due to promotion of the migration and proliferation of human hair follicle papilla cells. RNA sequencing is performed to explore the underlying mechanisms, which can activate ECM-receptor interaction pathway in favor of the wound healing process. The inhibiting effect of CNF@SITA@NZnS scaffold on DPP4+ fibroblasts can be a potential target to reduce scarring and promote skin regeneration.
Collapse
Affiliation(s)
- Xia Li
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Kai-Wen Zhang
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Zheng-Yu Zhang
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Zheng-Dong Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Feng-Lai Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China.
| | - Jinghua Chen
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China; Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
14
|
Liu J, Chen S, Zhang Z, Song X, Hou Z, Wang Z, Liu T, Yang L, Liu Y, Luo Z. The oxidized hyaluronic acid hydrogels containing paeoniflorin microspheres regulates the polarization of M1/M2 macrophages to promote wound healing. Int J Biol Macromol 2024; 282:137107. [PMID: 39515704 DOI: 10.1016/j.ijbiomac.2024.137107] [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: 07/12/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Controlling excessive inflammation of acute wound is an effective means to shorten the healing time. Therefore, targeted control of the inflammatory response of the wound is a promising therapeutic strategy. In this study, paeoniflorin (Pae) was encapsulated in microspheres and combined with oxidized hyaluronic acid hydrogels to prepare the hydrogel loaded with Pae microspheres (Pae-MPs@OHA) to promote the healing of acute wounds in rats. The results demonstrated that the particle size of the Pae-MPs was 6.84 ± 0.51 μm, and the positive charge was 26.87 ± 1.51 mV. The uniform spherical structure of the Pae-MPs was observed by TEM. The Pae-MPs@OHA can maintain colloidal state in the range of 0.1-3.16 Hz. FTIR suggested that Pae could be effectively wrapped in MPs, and SEM indicated that the Pae-MPs@OHA had a uniform network pore structure. The Pae-MPs@OHA can realize the sustained release of Pae for 96 h. Biocompatibility experiments showed that the Pae-MPs@OHA hydrogels were safe and available. The Pae-MPs@OHA hydrogels can accelerate wound healing in rats. HE and masson staining suggested that the Pae-MPs@OHA could reduce inflammatory cell infiltration, promote re-epithelialization and collagen formation. The Pae-MPs@OHA could decrease the number of M1 and increase the number of M2 in macrophages, thus regulating the release of inflammatory factor TNF-α and IL-1β. The results of molecular docking and western blot results also confirmed that the Pae-MPs@OHA could reduce the expression of NF-κB, pNF-κB, NLRP3, ASC and pro-caspase-1. These findings suggest that the Pae-MPs@OHA has great potential for application in the treatment of inflammatory wound.
Collapse
Affiliation(s)
- Jiarui Liu
- Shuren International College, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Siqi Chen
- School of Public Health, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Zijing Zhang
- Graduate School, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Xitong Song
- Graduate School, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Zhiquan Hou
- Graduate School, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Ziyi Wang
- Graduate School, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Tao Liu
- University of Michigan, Ann Arbor, School of Pharmacy, Integrated Pharmaceutical Sciences, 428 Church St, Ann Arbor, MI 48109, United States of America
| | - Liqun Yang
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yunen Liu
- Shuren International College, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China.
| | - Zhonghua Luo
- Shuren International College, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China.
| |
Collapse
|
15
|
Xiao F, Rui S, Zhang X, Ma Y, Wu X, Hao W, Huang G, Armstrong DG, Chen Q, Deng W. Accelerating diabetic wound healing with Ramulus Mori (Sangzhi) alkaloids via NRF2/HO-1/eNOS pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155990. [PMID: 39243750 DOI: 10.1016/j.phymed.2024.155990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/11/2024] [Accepted: 08/25/2024] [Indexed: 09/09/2024]
Abstract
Diabetic foot ulcers (DFUs) represent a severe complication of diabetes mellitus. Ramulus Mori (Sangzhi) alkaloids (SZ-A), an approved oral medication for type 2 diabetes, have not been explored for their potential to enhance the processes involved in diabetic wound healing. This study aims to investigate SZ-A's role in diabetic wound healing mechanisms. The in vivo experimentation involves dividing the subjects into NC and SZ-A groups, with SZ-A dosed at 200 and 400 mg/kg, to assess the therapeutic efficacy of SZ-A. The results of the animal studies show that SZ-A intervention accelerates the processes of diabetic angiogenesis and wound healing in a manner dependent on its concentration. Additionally, a pathological model using advanced glycation end products (AGEs) in HUVECs demonstrates SZ-A's cytoprotective effect. In vitro, SZ-A intervention significantly increases cell proliferation, migration and tube formation, protecting HUVECs from oxidative stress injury induced by AGEs. Mechanistically, SZ-A exerts a protective effect on HUVECs from oxidative stress damage through the activation of the NRF2/HO-1/eNOS signaling pathway. The findings suggest that SZ-A exhibits considerable potential as a promising candidate for treating DFUs, which will aid in more effectively integrating plant-based therapies into clinical settings.
Collapse
Affiliation(s)
- Fugang Xiao
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing University, Chongqing 400014, China
| | - Shunli Rui
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing University, Chongqing 400014, China
| | - Xiaoshi Zhang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing University, Chongqing 400014, China
| | - Yu Ma
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing University, Chongqing 400014, China
| | - Xiaohua Wu
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing University, Chongqing 400014, China
| | - Wei Hao
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing University, Chongqing 400014, China
| | - Guangbin Huang
- Department of Traumatology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing University, Chongqing 400014, China
| | - David G Armstrong
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Wuquan Deng
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Centre, Chongqing University, Chongqing 400014, China.
| |
Collapse
|
16
|
Eisenach IA, Lapii GA, Uzyumova AK, Lushnikova EL, Ovchinnikov VS, Solovieva AO, Naprimerov VA. Application of hs-CRP in Assessment of Tissue Inflammation Following Implantation of Biodegradable Polymer in Experiment. Int J Mol Sci 2024; 25:11183. [PMID: 39456966 PMCID: PMC11508534 DOI: 10.3390/ijms252011183] [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: 09/19/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Implants made of biodegradable polymers are replaced by regenerating tissues through inflammation. The changes occurring in tissues and the organism are of practical interest for studying the biocompatibility of the material and searching for systemic markers in the blood that reflect inflammation in peri-implantation tissues. The highly sensitive C-reactive protein (hs-CRP) measurements in blood and morphometric studies of tissue surrounding the implant were carried out in the experiment within three months of implantation of a biopolymer consisting of polycaprolactone (PCL) and polytrimethylene carbonate (PTMC). During the first month, tissue inflammation decreased, and the blood level of hs-CRP did not increase. The polymer biotransformation began in the tissues after a month of implantation and was accompanied by inflammation moving deeper into the matrix. Proliferation of inflammatory cells in tissues was reflected in an increase in the hs-CRP level three months after polymer installation. The result achieved confirmed the polymer's bioinertness. The level of hs-CRP in the blood of the animals correlated with the level of inflammation in peri-implantation tissues, reflecting the activity of inflammation in the process of polymer biotransformation. This inflammation protein can be recommended for assessing tissue processes following implantation of biopolymers and their biocompatibility.
Collapse
Affiliation(s)
- Igor A. Eisenach
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center for Fundamental and Translational Medicine, 2 Timakova St., Novosibirsk 630117, Russia; (G.A.L.); (A.K.U.); (E.L.L.)
| | - Galina A. Lapii
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center for Fundamental and Translational Medicine, 2 Timakova St., Novosibirsk 630117, Russia; (G.A.L.); (A.K.U.); (E.L.L.)
| | - Alexandra K. Uzyumova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center for Fundamental and Translational Medicine, 2 Timakova St., Novosibirsk 630117, Russia; (G.A.L.); (A.K.U.); (E.L.L.)
| | - Elena L. Lushnikova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center for Fundamental and Translational Medicine, 2 Timakova St., Novosibirsk 630117, Russia; (G.A.L.); (A.K.U.); (E.L.L.)
| | - Victor S. Ovchinnikov
- Research Institute of Clinical and Experimental Lymphology, Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 6 Arbuzova, Novosibirsk 630060, Russia; (V.S.O.); (A.O.S.)
| | - Anastasia O. Solovieva
- Research Institute of Clinical and Experimental Lymphology, Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 6 Arbuzova, Novosibirsk 630060, Russia; (V.S.O.); (A.O.S.)
| | - Vasiliy A. Naprimerov
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Prospekt Lavrentyeva, Novosibirsk 630090, Russia;
| |
Collapse
|
17
|
Wang M, Hong Y, Fu X, Sun X. Advances and applications of biomimetic biomaterials for endogenous skin regeneration. Bioact Mater 2024; 39:492-520. [PMID: 38883311 PMCID: PMC11179177 DOI: 10.1016/j.bioactmat.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 06/18/2024] Open
Abstract
Endogenous regeneration is becoming an increasingly important strategy for wound healing as it facilitates skin's own regenerative potential for self-healing, thereby avoiding the risks of immune rejection and exogenous infection. However, currently applied biomaterials for inducing endogenous skin regeneration are simplistic in their structure and function, lacking the ability to accurately mimic the intricate tissue structure and regulate the disordered microenvironment. Novel biomimetic biomaterials with precise structure, chemical composition, and biophysical properties offer a promising avenue for achieving perfect endogenous skin regeneration. Here, we outline the recent advances in biomimetic materials induced endogenous skin regeneration from the aspects of structural and functional mimicry, physiological process regulation, and biophysical property design. Furthermore, novel techniques including in situ reprograming, flexible electronic skin, artificial intelligence, single-cell sequencing, and spatial transcriptomics, which have potential to contribute to the development of biomimetic biomaterials are highlighted. Finally, the prospects and challenges of further research and application of biomimetic biomaterials are discussed. This review provides reference to address the clinical problems of rapid and high-quality skin regeneration.
Collapse
Affiliation(s)
- Mengyang Wang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
| | - Yiyue Hong
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| |
Collapse
|
18
|
Li X, An T, Yang Y, Xu Z, Chen S, Yi Z, Deng C, Zhou F, Man Y, Hu C. TLR9 activation in large wound induces tissue repair and hair follicle regeneration via γδT cells. Cell Death Dis 2024; 15:598. [PMID: 39153998 PMCID: PMC11330466 DOI: 10.1038/s41419-024-06994-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
The mechanisms underlying tissue repair in response to damage have been one of main subjects of investigation. Here we leverage the wound-induced hair neogenesis (WIHN) models in adult mice to explore the correlation between degree of damage and the healing process and outcome. The multimodal analysis, in combination with single-cell RNA sequencing help to explore the difference in wounds of gentle and heavy damage degrees, identifying the potential role of toll-like receptor 9 (TLR9) in sensing the injury and regulating the immune reaction by promoting the migration of γδT cells. The TLR9 deficient mice or wounds injected with TLR9 antagonist have greatly impaired healing and lower WIHN levels. Inhibiting the migration of γδT cells or knockout of γδT cells also suppress the wound healing and regeneration, which can't be rescued by TLR9agonist. Finally, the amphiregulin (AREG) is shown as one of most important effectors secreted by γδT cells and keratinocytes both in silicon or in the laboratory, whose expression influences WIHN levels and the expression of stem cell markers. In total, our findings reveal a previously unrecognized role for TLR9 in sensing skin injury and influencing the tissue repair and regeneration by modulation of the migration of γδT cells, and identify the TLR9-γδT cells-areg axis as new potential targets for enhancing tissue regeneration.
Collapse
Affiliation(s)
- Xinhui Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Tiantian An
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yang Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhaoyu Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shuaidong Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zumu Yi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen Deng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Feng Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Man
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Chen Hu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
19
|
Li N, Cao Y, Liu J, Zou W, Chen M, Cao H, Deng S, Liang J, Yuan T, Wang Q, Fan Y, Zhang X. Microenvironment-responsive release of Mg 2+ from tannic acid decorated and multilevel crosslinked hydrogels accelerates infected wound healing. J Mater Chem B 2024; 12:6856-6873. [PMID: 38904349 DOI: 10.1039/d4tb01000k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
The management of chronic infected wounds poses significant challenges due to frequent bacterial infections, high concentrations of reactive oxygen species, abnormal immune regulation, and impaired angiogenesis. This study introduces a novel, microenvironment-responsive, dual dynamic, and covalently bonded hydrogel, termed OHA-P-TA/G/Mg2+. It is derived from the reaction of tannic acid (TA) with phenylboronic acids (PBA), which are grafted onto oxidized hyaluronic acid (OHA-P-TA), combined with GelMA (G) via a Schiff base and chemical bonds, along with the incorporation of Mg2+. This hydrogel exhibits pH and ROS dual-responsiveness, demonstrating effective antibacterial capacity, antioxidant ability, and the anti-inflammatory ability under distinct acidic and oxidative microenvironments. Furthermore, the release of Mg2+ from the TA-Mg2+ network (TA@Mg2+) promotes the transformation of pro-inflammatory M1 phenotype macrophages to anti-inflammatory M2 phenotype, showing a microenvironment-responsive response. Finally, in vivo results indicate that the OHA-P-TA/G/Mg2+ hydrogel enhances epithelial regeneration, collagen deposition, and neovascularization, showing great potential as an effective dressing for infected wound repair.
Collapse
Affiliation(s)
- Na Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
| | - Yi Cao
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Jingyi Liu
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Wen Zou
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
| | - Manyu Chen
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Hongfu Cao
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Siyan Deng
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
| | - Tun Yuan
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China.
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, China
| |
Collapse
|
20
|
Yuan Y, Zhao H, Yin X, Wang D, Mei X, Zhang P. Alloy nanozyme-reinforced hyaluronic acid-based hydrogel with wound environment-responsive properties for synergistically accelerating infectious wound healing. Int J Biol Macromol 2024; 269:131896. [PMID: 38677681 DOI: 10.1016/j.ijbiomac.2024.131896] [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: 01/18/2024] [Revised: 03/12/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
The recovery of infectious wound tissues presents a significant global health challenge due to the impediments posed by the harsh healing microenvironment, which includes ongoing bacterial invasion, high oxidative stress, inflammatory response, and impaired angiogenesis. To overcome the above issues, we propose a composite hydrogel based on the multiple-crosslinked mechanism involving the covalent network of CC bonds within catechol and maleic-modified HA (CMHA), the self-assembly network of glycyrrhizic acid (GA), and the metal-polyphenol coordination induced by ZHMCe for accelerating infectious wound healing. The resulting CMHA/GA/ZHMCe hydrogels demonstrate enhanced mechanical, adhesive, antioxidative, and antibacterial properties. Importantly, the hydrogel system possesses wound environment-responsive properties that allow it to adapt to the specific therapeutic requirements of different stages by regulating various enzyme activities in the healing of infected wounds. Furthermore, the biocompatible CMHA/GA/ZHMCe shows the ability to promote cell migration and angiogenesis in vitro while reprogramming macrophages toward an anti-inflammatory phenotype due to the effective release of active ingredients. In vivo experiments confirm that the CMHA/GA/ZHMCe hydrogel significantly enhances infectious wound healing by accelerating re-epithelialization, promoting collagen deposition, regulating inflammation, and contributing to vascularization. These findings underscore the therapeutic potential of our hydrogel dressings for the treatment of bacterially infected cutaneous wound healing.
Collapse
Affiliation(s)
- Yajiang Yuan
- Department of Orthopedic, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Haosen Zhao
- Department of Orthopedic, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Xuechen Yin
- Department of Laboratory Medicine, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Dahao Wang
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121002, China
| | - Xifan Mei
- Department of Orthopedic, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China.
| | - Peng Zhang
- Department of Orthopedic, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China.
| |
Collapse
|
21
|
Piatnitskaia S, Rafikova G, Bilyalov A, Chugunov S, Akhatov I, Pavlov V, Kzhyshkowska J. Modelling of macrophage responses to biomaterials in vitro: state-of-the-art and the need for the improvement. Front Immunol 2024; 15:1349461. [PMID: 38596667 PMCID: PMC11002093 DOI: 10.3389/fimmu.2024.1349461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/21/2024] [Indexed: 04/11/2024] Open
Abstract
The increasing use of medical implants in various areas of medicine, particularly in orthopedic surgery, oncology, cardiology and dentistry, displayed the limitations in long-term integration of available biomaterials. The effective functioning and successful integration of implants requires not only technical excellence of materials but also consideration of the dynamics of biomaterial interaction with the immune system throughout the entire duration of implant use. The acute as well as long-term decisions about the efficiency of implant integration are done by local resident tissue macrophages and monocyte-derived macrophages that start to be recruited during tissue damage, when implant is installed, and are continuously recruited during the healing phase. Our review summarized the knowledge about the currently used macrophages-based in vitro cells system that include murine and human cells lines and primary ex vivo differentiated macrophages. We provided the information about most frequently examined biomarkers for acute inflammation, chronic inflammation, foreign body response and fibrosis, indicating the benefits and limitations of the model systems. Particular attention is given to the scavenging function of macrophages that controls dynamic composition of peri-implant microenvironment and ensures timely clearance of microorganisms, cytokines, metabolites, extracellular matrix components, dying cells as well as implant debris. We outline the perspective for the application of 3D systems for modelling implant interaction with the immune system in human tissue-specific microenvironment avoiding animal experimentation.
Collapse
Affiliation(s)
- Svetlana Piatnitskaia
- Cell Technology Laboratory, Institute of Fundamental Medicine, Bashkir State Medical University, Ufa, Russia
| | - Guzel Rafikova
- Additive Technology Laboratory, Institute of Fundamental Medicine, Bashkir State Medical University, Ufa, Russia
- Laboratory of Immunology, Institute of Urology and Clinical Oncology, Bashkir State Medical University, Ufa, Russia
| | - Azat Bilyalov
- Additive Technology Laboratory, Institute of Fundamental Medicine, Bashkir State Medical University, Ufa, Russia
| | - Svyatoslav Chugunov
- Additive Technology Laboratory, Institute of Fundamental Medicine, Bashkir State Medical University, Ufa, Russia
| | - Iskander Akhatov
- Laboratory of Mathematical modeling, Institute of Fundamental Medicine, Bashkir State Medical University, Ufa, Russia
| | - Valentin Pavlov
- Institute of Urology and Clinical Oncology, Department of Urology, Bashkir State Medical University, Ufa, Russia
| | - Julia Kzhyshkowska
- Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, Mannheim, Germany
| |
Collapse
|