1
|
Zhang SJ, Xu R, He SB, Sun R, Wang GN, Wei SY, Yan XY, Fan KL. Nanozyme-driven multifunctional dressings: moving beyond enzyme-like catalysis in chronic wound treatment. Mil Med Res 2025; 12:27. [PMID: 40448212 DOI: 10.1186/s40779-025-00611-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/26/2025] [Indexed: 06/02/2025] Open
Abstract
The treatment of chronic wounds presents significant challenges due to the necessity of accelerating healing within complex microenvironments characterized by persistent inflammation and biochemical imbalances. Factors such as bacterial infections, hyperglycemia, and oxidative stress disrupt cellular functions and impair angiogenesis, substantially delaying wound repair. Nanozymes, which are engineered nanoscale materials with enzyme-like activities, offer distinct advantages over conventional enzymes and traditional nanomaterials, making them promising candidates for chronic wound treatment. To enhance their clinical potential, nanozyme-based catalytic systems are currently being optimized through formulation advancements and preclinical studies assessing their biocompatibility, anti-oxidant activity, antibacterial efficacy, and tissue repair capabilities, ensuring their safety and clinical applicability. When integrated into multifunctional wound dressings, nanozymes modulate reactive oxygen species levels, promote tissue regeneration, and simultaneously combat infections and oxidative damage, extending beyond conventional enzyme-like catalysis in chronic wound treatment. The customizable architectures of nanozymes enable precise therapeutic applications, enhancing their effectiveness in managing complex wound conditions. This review provides a comprehensive analysis of the incorporation of nanozymes into wound dressings, detailing fabrication methods and emphasizing their transformative potential in chronic wound management. By identifying and addressing key limitations, we introduce strategic advancements to drive the development of nanozyme-driven dressings, paving the way for next-generation chronic wound treatments.
Collapse
Affiliation(s)
- Si-Jie Zhang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Ran Xu
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Shao-Bin He
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Laboratory of Clinical Pharmacy, Department of Pharmacy, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Rong Sun
- Department of Radiation Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Guan-Nan Wang
- Shenyang Key Laboratory of Medical Molecular Theranostic Probes in School of Pharmacy, Shenyang Medical College, Shenyang, 110034, China
| | - Shu-Yi Wei
- Peking University People's Hospital, Beijing, 100044, China
| | - Xi-Yun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China
| | - Ke-Long Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China.
| |
Collapse
|
2
|
Zhu K, Yao Z, Gu T, Jiang X, Zhou J, Li D. Study of the ability of polysaccharides isolated from Zizania latifolia to promote wound healing in mice via in vitro screening and in vivo evaluation. Food Chem 2025; 464:141810. [PMID: 39532060 DOI: 10.1016/j.foodchem.2024.141810] [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: 08/02/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Swollen culms of Zizania latifolia possess a peculiar flavor, nutritional value and health-promoting effects. This study investigated the wound healing ability of polysaccharides isolated from Zizania latifolia (ZLP). Through in vitro assays, ZLP demonstrated negligible cytotoxicity and promoted the migration and phagocytosis of Staphylococcus aureus by macrophages where ZLP-2 had a significantly greater effect. Moreover, ZLP-2 has a higher purity and molecular weight, an integral and large lamellar morphology, and a larger particle size in solution, suggesting its ability to form films. Thus, ZLP-2 was fabricated as a gel formulation for topical application. The in vivo results showed that the ZLP-2 gel had significantly greater effects than did the autonomous healing process according to quantitative analysis of the wound closure area. The ZLP-2 gel decreased the levels of TNF-α and IL-6 in wounded tissue. Furthermore, the quantification of the molecular indexes via qRT-PCR revealed that the levels of tnf-α, tgf-β, mcp-1, ifn-γ, inos, and vegf in the treated group increased during the inflammatory and proliferation stages but decreased after wound closure. These results suggest that ZLP-2 has great potential in pharmaceutical applications for wound care.
Collapse
Affiliation(s)
- Kehan Zhu
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China
| | - Zhen Yao
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China
| | - Tianyi Gu
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China
| | - Xianhao Jiang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China
| | - Jianqin Zhou
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China.
| | - Duxin Li
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
3
|
Chen C, Chen Y, Ye Z, Ali A, Yao S. Bioactive Deep Eutectic Solvent-Involved Sprayable Versatile Hydrogel for Monkeypox Virus Lesions Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:2148-2168. [PMID: 39727382 DOI: 10.1021/acsami.4c14905] [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: 12/28/2024]
Abstract
To address the issues of infectious virus, bacterial secondary infections, skin pigmentation, and scarring caused by monkeypox virus (MPXV), a sprayable hydrogel with versatile functions was developed with comprehensive properties. Based on current research, the bioactive deep eutectic solvent (DES) of rosmarinic acid-proanthocyanidin-glycol (RPG) was designed and synthesized as active agent, and molecular docking was applied to discover its binding to MPXV proteins through H-bonds and van der Waals interactions, and the docking results show the binding energies between RA, PC, Gly and MPXV proteins are -58.7188, -50.2311, and -18.4755 kcal/mol, respectively. Additionally, poly(vinyl alcohol) (PVA), borate, and xylitol (Xyl) were integrated with RPG to prepare the PB-RPG-Xyl hydrogel, which was characterized by popular ways. The pH-responsive properties of the hydrogel accelerated the release of RPG under acidic conditions, resulting in an increased cumulative release percentage of 84.83% at pH 5.5 at 210 min. Besides that, it was proved to have the expected sprayability, self-healing, adhesion, and shape-adaptability. The results of molecular dynamic simulation were meaningful to understanding its formation and self-healing mechanisms. Furthermore, the hydrogel shows ideal degradability, removability, and biocompatibility. Lastly, its multiple functions were systematically explored, including UV-blocking, blood clotting, cooling, antioxidant, antibacterial, and virus inhibition properties. The developed sprayable PB-RPG-Xyl hydrogel represents the first promising dressing based on natural bioactive DES for MPXV lesions management, which not only expands the application of green solvents in health care but also provides a convenient and effective treatment process for MPXV infection in the face of difficult skin lesions and complex treatment needs.
Collapse
Affiliation(s)
- Chen Chen
- School of Chemical Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yu Chen
- South Sichuan Institute of Translational Medicine, College of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Zhiyi Ye
- School of Chemical Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Ahmad Ali
- School of Chemical Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Shun Yao
- School of Chemical Engineering, Sichuan University, Chengdu 610065, P. R. China
| |
Collapse
|
4
|
Cao B, Da X, Wu W, Xie J, Li X, Wang X, Xu H, Gao J, Yang H, Su J. Multifunctional human serum albumin-crosslinked and self-assembling nanoparticles for therapy of periodontitis by anti-oxidation, anti-inflammation and osteogenesis. Mater Today Bio 2024; 28:101163. [PMID: 39183771 PMCID: PMC11341939 DOI: 10.1016/j.mtbio.2024.101163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease that can result in the irreversible loss of tooth-supporting tissues and elevate the likelihood and intensity of systemic diseases. The presence of reactive oxygen species (ROS) and associated related oxidative stress is intricately linked to the progression and severity of periodontal inflammation. Targeted removal of local ROS may serve to attenuate inflammation, improve the unfavorable periodontal microenvironment and potentially reverse ensuing pathological cascades. These ROS scavenging nanoparticles, which possess additional characteristics such as anti-inflammation and osteogenic differentiation, are highly sought after for the treatment of periodontitis. In this study, negative charged human serum albumin-crosslinked manganese-doped self-assembling Prussian blue nanoparticles (HSA-MDSPB NPs) were fabricated. These nanoparticles demonstrate the ability to scavenge multiple ROS including superoxide anion, free hydroxyl radicals, singlet oxygen and hydrogen peroxide. Additionally, HSA-MDSPB NPs exhibit the capacity to alleviate inflammation in gingiva and alveolar bone both in vitro and in vivo. Furthermore, HSA-MDSPB NPs have been shown to play a role in promoting the polarization of macrophages from the M1 to M2 phenotype, resulting in reduced production of pro-inflammatory cytokines. More attractively, HSA-MDSPB NPs have been demonstrated to enhance cellular osteogenic differentiation. These properties of HSA-MDSPB NPs contribute to decreased inflammation, extracellular matrix degradation and bone loss in periodontal tissue. In conclusion, the multifunctional nature of HSA-MDSPB NPs provides a promising therapeutic approach for the treatment of periodontitis.
Collapse
Affiliation(s)
- Bangping Cao
- 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, China
| | - Xuanbo Da
- Department of General Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710000, China
| | - Wenjing Wu
- 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, China
| | - Jian Xie
- 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, 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, China
| | - Xin Wang
- 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, China
| | - Hui Xu
- 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, China
| | - Jianfang Gao
- 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, China
| | - Hui Yang
- 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, 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, China
| |
Collapse
|
5
|
Zhang M, Chen S, Xu G, Lu W, Li J, Zhang J, Zhang Z, Zhu J, Pan X. Ultra-Fast Selenol-Yne Click (SYC) Reaction Enables Poly(selenoacetal) Covalent Adaptable Network Formation. Angew Chem Int Ed Engl 2024; 63:e202410245. [PMID: 38887146 DOI: 10.1002/anie.202410245] [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: 05/30/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 06/20/2024]
Abstract
The emergence of covalent adaptable networks (CANs) based on dynamic covalent bonds (DCBs) presents a promising avenue for achieving resource recovery and utilization. In this study, we discovered a dynamic covalent bond called selenacetal, which is obtained through a double click reaction between selenol and activated alkynes. Density functional theory (DFT) calculations demonstrated that the ΔG for the formation of selenoacetals ranges from 12 to 18 kJ mol-1, suggesting its potential for dynamic reversibility. Dynamic exchange experiments involving small molecules and polymers provide substantial evidence supporting the dynamic exchange properties of selenoacetals. By utilizing this highly efficient click reaction, we successfully synthesized dynamic materials based on selenoacetal with remarkable reprocessing capabilities without any catalysts. These materials exhibit chemical recycling under alkaline conditions, wherein selenoacetal (SA) can decompose into active enone selenide (ES) and diselenides. Reintroducing selenol initiates a renewed reaction with the enone selenide, facilitating material recycling and yielding a newly developed dynamic material exhibiting both photo- and thermal responsiveness. The results underscore the potential of selenoacetal polymers in terms of recyclability and selective degradation, making them a valuable addition to conventional covalent adaptable networks.
Collapse
Affiliation(s)
- Mengyao Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Department Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Sisi Chen
- State and Local Joint Engineering Laboratory for Novel Functional Department Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Guichuan Xu
- State and Local Joint Engineering Laboratory for Novel Functional Department Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Weihong Lu
- State and Local Joint Engineering Laboratory for Novel Functional Department Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Jiajia Li
- State and Local Joint Engineering Laboratory for Novel Functional Department Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Jiandong Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Department Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Zhengbiao Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Department Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Jian Zhu
- State and Local Joint Engineering Laboratory for Novel Functional Department Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Xiangqiang Pan
- State and Local Joint Engineering Laboratory for Novel Functional Department Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| |
Collapse
|
6
|
Lai Y, Jiang M, Zhang X, Zhang L, Chen Z, Du Y, Wang S, Zhao J, Li Z. Novel endoscopic tattooing dye based on polyvinylpyrrolidone-modified polydopamine nanoparticles for labeling gastrointestinal lesions. J Mater Chem B 2024; 12:9345-9356. [PMID: 39171740 DOI: 10.1039/d4tb01298d] [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: 08/23/2024]
Abstract
Endoscopic tattooing is a localization technique that is particularly important for identifying gastrointestinal lesions for follow-up and subsequent treatment. However, the dyes currently used for endoscopic tattooing have a short tattooing time, high cost, and many side effects. Herein, we designed and prepared polydopamine (PDA) nanoparticles modified with polyvinylpyrrolidone (PVP) for endoscopic tattooing using a physical encapsulation method. PDA has good stability and high adhesion properties, and its stability was further enhanced after PVP modification. In vitro and in vivo tests demonstrated that PDA/PVP has good biosafety. Endoscopic tattooing with PDA/PVP in a porcine model showed that the dye could be stabilized in the digestive tract for at least 60 days. Furthermore, our research results demonstrated that PDA/PVP has excellent reactive oxygen species (ROS) and reactive nitrogen species (RNS) scavenging ability and can promote wound healing. Overall, the strategy proposed herein will lead to the use of an innovative dye for endoscopic tattooing of gastrointestinal lesions.
Collapse
Affiliation(s)
- Yongkang Lai
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
- Department of Gastroenterology, Ganzhou People's Hospital, Jiangxi Medical College, Nanchang University, Ganzhou 341000, China
| | - Mengni Jiang
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Xinyuan Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Liang Zhang
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Zheng Chen
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yiqi Du
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital; National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, China.
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Jiulong Zhao
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Zhaoshen Li
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
7
|
Zhou M, Yuan T, Shang L. 3D Printing of Naturally Derived Adhesive Hemostatic Sponge. RESEARCH (WASHINGTON, D.C.) 2024; 7:0446. [PMID: 39119591 PMCID: PMC11309851 DOI: 10.34133/research.0446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024]
Abstract
Hydrogel hemostatic sponges have been recognized for its effectiveness in wound treatment due to its excellent biocompatibility, degradability, as well as multi-facet functionalities. Current research focuses on optimizing the composition and structure of the sponge to enhance its therapeutic effectiveness. Here, we propose an adhesive hydrogel made from purely natural substances extracted from okra and Panax notoginseng. We utilize 3-dimensional (3D) printing technology to fabricate the hemostatic hydrogel scaffold, incorporating gelatin into the hydrogel and refining the mixing ratio. The interaction between gelatin and okra polyphenols contributes to successful injectability as well as stability of the printed scaffold. The okra in the scaffold exhibits favorable adhesion and hemostatic effects, and the total saponins of Panax notoginseng facilitate angiogenesis. Through in vitro experiments, we have substantiated the scaffold's excellent stability, adhesion, biocompatibility, and angiogenesis-promoting ability. Furthermore, in vivo experiments have demonstrated its dual functionality in rapid hemostasis and wound repair. These features suggest that the 3D-printed, natural substance-derived hydrogel scaffolds have valuable potential in wound healing and related applications.
Collapse
Affiliation(s)
- Minyu Zhou
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Tao Yuan
- Department of Spine Surgery,
Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Luoran Shang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences,
Fudan University, Shanghai, China
| |
Collapse
|
8
|
Xiao X, Zhao F, DuBois DB, Liu Q, Zhang YL, Yao Q, Zhang GJ, Chen S. Nanozymes for the Therapeutic Treatment of Diabetic Foot Ulcers. ACS Biomater Sci Eng 2024; 10:4195-4226. [PMID: 38752382 DOI: 10.1021/acsbiomaterials.4c00470] [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: 07/09/2024]
Abstract
Diabetic foot ulcers (DFU) are chronic, refractory wounds caused by diabetic neuropathy, vascular disease, and bacterial infection, and have become one of the most serious and persistent complications of diabetes mellitus because of their high incidence and difficulty in healing. Its malignancy results from a complex microenvironment that includes a series of unfriendly physiological states secondary to hyperglycemia, such as recurrent infections, excessive oxidative stress, persistent inflammation, and ischemia and hypoxia. However, current common clinical treatments, such as antibiotic therapy, insulin therapy, surgical debridement, and conventional wound dressings all have drawbacks, and suboptimal outcomes exacerbate the financial and physical burdens of diabetic patients. Therefore, development of new, effective and affordable treatments for DFU represents a top priority to improve the quality of life of diabetic patients. In recent years, nanozymes-based diabetic wound therapy systems have been attracting extensive interest by integrating the unique advantages of nanomaterials and natural enzymes. Compared with natural enzymes, nanozymes possess more stable catalytic activity, lower production cost and greater maneuverability. Remarkably, many nanozymes possess multienzyme activities that can cascade multiple enzyme-catalyzed reactions simultaneously throughout the recovery process of DFU. Additionally, their favorable photothermal-acoustic properties can be exploited for further enhancement of the therapeutic effects. In this review we first describe the characteristic pathological microenvironment of DFU, then discuss the therapeutic mechanisms and applications of nanozymes in DFU healing, and finally, highlight the challenges and perspectives of nanozyme development for DFU treatment.
Collapse
Affiliation(s)
- Xueqian Xiao
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
| | - Fei Zhao
- Institute of Hematology, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430065, China
| | - Davida Briana DuBois
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, California 95064, United States
| | - Qiming Liu
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, California 95064, United States
| | - Yu Lin Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
- Hubei Shizhen Laboratory, Wuhan, Hubei 430065, China
| | - Qunfeng Yao
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
- Hubei Shizhen Laboratory, Wuhan, Hubei 430065, China
| | - Guo-Jun Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
- Hubei Shizhen Laboratory, Wuhan, Hubei 430065, China
| | - Shaowei Chen
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, California 95064, United States
| |
Collapse
|
9
|
Liu WS, Chen Z, Lu ZM, Dong JH, Wu JH, Gao J, Deng D, Li M. Multifunctional hydrogels based on photothermal therapy: A prospective platform for the postoperative management of melanoma. J Control Release 2024; 371:406-428. [PMID: 38849093 DOI: 10.1016/j.jconrel.2024.06.001] [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: 12/20/2023] [Revised: 03/22/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
Preventing the recurrence of melanoma after surgery and accelerating wound healing are among the most challenging aspects of melanoma management. Photothermal therapy has been widely used to treat tumors and bacterial infections and promote wound healing. Owing to its efficacy and specificity, it may be used for postoperative management of tumors. However, its use is limited by the uncontrollable distribution of photosensitizers and the likelihood of damage to the surrounding normal tissue. Hydrogels provide a moist environment with strong biocompatibility and adhesion for wound healing owing to their highly hydrophilic three-dimensional network structure. In addition, these materials serve as excellent drug carriers for tumor treatment and wound healing. It is possible to combine the advantages of both of these agents through different loading modalities to provide a powerful platform for the prevention of tumor recurrence and wound healing. This review summarizes the design strategies, research progress and mechanism of action of hydrogels used in photothermal therapy and discusses their role in preventing tumor recurrence and accelerating wound healing. These findings provide valuable insights into the postoperative management of melanoma and may guide the development of promising multifunctional hydrogels for photothermal therapy.
Collapse
Affiliation(s)
- Wen-Shang Liu
- Department of Dermatology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, People's Republic of China
| | - Zhuo Chen
- Department of Dermatology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, People's Republic of China
| | - Zheng-Mao Lu
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, People's Republic of China
| | - Jin-Hua Dong
- Women and Children Hospital Affiliated to Jiaxing University, 2468 Middle Ring Eastern Road, Jiaxing City, Zhejiang 314000, People's Republic of China
| | - Jin-Hui Wu
- Ophthalmology Department of the Third Affiliated Hospital of Naval Medical University, Shanghai 201805, People's Republic of China.
| | - Jie Gao
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai 200433, People's Republic of China.
| | - Dan Deng
- Department of Dermatology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, People's Republic of China.
| | - Meng Li
- Department of Dermatology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, People's Republic of China.
| |
Collapse
|
10
|
Cheng L, Zhang S, Zhang Q, Gao W, Wang B, Mu S. Fabrication of pH-stimuli hydrogel as bioactive materials for wound healing applications. Heliyon 2024; 10:e32864. [PMID: 39021919 PMCID: PMC11252711 DOI: 10.1016/j.heliyon.2024.e32864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024] Open
Abstract
Hydrogels exhibit exceptional suitability as wound dressing due to their remarkable three-dimensional (3D) characteristics. Here, we have reported the fabrication of hydrogels from biopolymers carboxymethyl cellulose (CMC), polyvinyl alcohol (PVA), and gelatin via a simple blending method to mimic the natural extracellular matrix. Scanning electron microscopy (SEM), water contact meters (WCM), and Fourier-transform infrared spectroscopy (FTIR) were used to evaluate the chemical structural, morphological, and wettability behavior. The wetting and degradation behavior were also found to be different for different formulations (Min. (51.60o) and Max. (113.60o)) and (Min. (38.82 mg) and Max. (3.72 mg)), respectively. Swelling was investigated in different media, including phosphate buffer saline solution (PBS) and aqueous media. It was observed that the hydrogel displayed the highest degree of swelling in an aqueous medium (Min. (597.32-1121.49 %) and Max. (1089.51-2139.73 %)) compared to PBS media (Min. (567.01-1021.85 %) and Max. (899.13-1639.17 %)). The release of Neomycin was studied in a PBS medium via the Franz diffusion method at 37 °C. The maximal release in various media demonstrated pH-responsive behavior. The viability and proliferation of fibroblast (3T3) cell lines were examined in vitro to evaluate cytocompatibility. Human Embryonic Kidney (HEK) 293 cells were used to evaluate the hydrogels' ability to promote vascularization and angiogenesis. Therefore, the data demonstrate that hydrogels that have been manufactured have qualities that make them promising for use as wound dressings in wound healing applications.
Collapse
Affiliation(s)
- Liang Cheng
- Department of Burns and Plastic Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Song Zhang
- Department of Burns and Plastic Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Qian Zhang
- Department of Burns and Plastic Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Wenjie Gao
- Department of Burns and Plastic Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Benfeng Wang
- Department of Burns and Plastic Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Shengzhi Mu
- Department of Burns and Plastic Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| |
Collapse
|
11
|
Liu Q, Wang C, Cheng M, Hu L, Zhang Z, Sun Q, Wang S, Fan Y, Pan P, Chen J. Self-Healing Conductive Hydrogels with Dynamic Dual Network Structure Accelerate Infected Wound Healing via Photothermal Antimicrobial and Regulating Inflammatory Response. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30776-30792. [PMID: 38848491 DOI: 10.1021/acsami.4c04113] [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: 06/09/2024]
Abstract
Wound infections are an escalating clinical challenge with continuous inflammatory response and the threat of drug-resistant bacteria. Herein, a series of self-healing conductive hydrogels were designed based on carboxymethyl chitosan/oxidized sodium alginate/polymerized gallic acid/Fe3+ (CMC/OSA/pGA/Fe3+, COGFe) for promoting infected wound healing. The Schiff base and catechol-Fe3+ chelation in the dynamical dual network structure of the hydrogels endowed dressings with good toughness, conductivity, adhesion, and self-healing properties, thus flexibly adapting to the deformation of skin wounds. In terms of ultraviolet (UV) resistance and scavenging of reactive oxygen species (ROS), the hydrogels significantly reduced oxidative stress at the wound site. Additionally, the hydrogels with photothermal therapy (PTT) achieved a 95% bactericidal rate in 5 min of near-infrared (NIR) light radiation by disrupting the bacterial cell membrane structure through elevated temperature. Meanwhile, the inherent antimicrobial properties of GA could reduce healthy tissue damage caused by excessive heat. The composite hydrogels could effectively promote the proliferation and migration of fibroblasts and possess good biocompatibility and hemostatic effect. In full-thickness infected wound repair experiments in rats, the COGFe5 hydrogel combined with NIR effectively killed bacteria, modulated macrophage polarization (M1 to M2 phenotype) to improve the immune microenvironment of the wound, and shortened the repair time by accelerating the expression of collagen deposition (TGF-β) and vascular factors (CD31). This combined therapy might provide a prospective strategy for infectious wound treatment.
Collapse
Affiliation(s)
- Qing Liu
- Marine College, Shandong University, Weihai 264209, China
| | - Chunxiao Wang
- Marine College, Shandong University, Weihai 264209, China
| | - Meiqi Cheng
- Marine College, Shandong University, Weihai 264209, China
| | - Le Hu
- Marine College, Shandong University, Weihai 264209, China
| | - Ziyue Zhang
- Marine College, Shandong University, Weihai 264209, China
| | - Qisen Sun
- Marine College, Shandong University, Weihai 264209, China
| | - Shaoshen Wang
- Marine College, Shandong University, Weihai 264209, China
| | - Yinuo Fan
- Marine College, Shandong University, Weihai 264209, China
| | - Panpan Pan
- Marine College, Shandong University, Weihai 264209, China
| | - Jingdi Chen
- Marine College, Shandong University, Weihai 264209, China
| |
Collapse
|
12
|
Wang Y, Zhang H, Qiang H, Li M, Cai Y, Zhou X, Xu Y, Yan Z, Dong J, Gao Y, Pan C, Yin X, Gao J, Zhang T, Yu Z. Innovative Biomaterials for Bone Tumor Treatment and Regeneration: Tackling Postoperative Challenges and Charting the Path Forward. Adv Healthc Mater 2024; 13:e2304060. [PMID: 38429938 DOI: 10.1002/adhm.202304060] [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: 11/19/2023] [Revised: 02/24/2024] [Indexed: 03/03/2024]
Abstract
Surgical resection of bone tumors is the primary approach employed in the treatment of bone cancer. Simultaneously, perioperative interventions, particularly postoperative adjuvant anticancer strategies, play a crucial role in achieving satisfactory therapeutic outcomes. However, the occurrence of postoperative bone tumor recurrence, metastasis, extensive bone defects, and infection are significant risks that can result in unfavorable prognoses or even treatment failure. In recent years, there has been significant progress in the development of biomaterials, leading to the emergence of new treatment options for bone tumor therapy and bone regeneration. This progress report aims to comprehensively analyze the strategic development of unique therapeutic biomaterials with inherent healing properties and bioactive capabilities for bone tissue regeneration. These composite biomaterials, classified into metallic, inorganic non-metallic, and organic types, are thoroughly investigated for their responses to external stimuli such as light or magnetic fields, internal interventions including chemotherapy or catalytic therapy, and combination therapy, as well as their role in bone regeneration. Additionally, an overview of self-healing materials for osteogenesis is provided and their potential applications in combating osteosarcoma and promoting bone formation are explored. Furthermore, the safety concerns of integrated materials and current limitations are addressed, while also discussing the challenges and future prospects.
Collapse
Affiliation(s)
- Yu Wang
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, P. R. China
| | - Huaiyuan Zhang
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, P. R. China
| | - Huifen Qiang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, P. R. China
| | - Meigui Li
- School of Pharmacy, Henan University, Kaifeng City, Henan, 475004, P. R. China
| | - Yili Cai
- Department of Gastroenterology, Naval Medical Center, Naval Medical University, Shanghai, 200052, P. R. China
| | - Xuan Zhou
- School of Pharmacy, Henan University, Kaifeng City, Henan, 475004, P. R. China
| | - Yanlong Xu
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, P. R. China
| | - Zhenzhen Yan
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
| | - Jinhua Dong
- The Women and Children Hospital Affiliated to Jiaxing University, Jiaxing, Zhejiang, 314000, P. R. China
| | - Yuan Gao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200433, P. R. China
| | - Chengye Pan
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
| | - Xiaojing Yin
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
| | - Jie Gao
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, P. R. China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, P. R. China
| | - Zuochong Yu
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, P. R. China
| |
Collapse
|
13
|
Wu Y, Bei Y, Li W, Lu W, Zhu J, Zhang Z, Zhang T, Liu S, Chen K, Jin H, Li L, Li M, Gao J, Pan X. Advanced Multifunctional Hydrogels for Enhanced Wound Healing through Ultra-Fast Selenol-S NAr Chemistry. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400898. [PMID: 38647422 DOI: 10.1002/advs.202400898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/26/2024] [Indexed: 04/25/2024]
Abstract
Fabrication of versatile hydrogels in a facile and effective manner represents a pivotal challenge in the field of biomaterials. Herein, a novel strategy is presented for preparing on-demand degradable hydrogels with multilevel responsiveness. By employing selenol-dichlorotetrazine nucleophilic aromatic substitution (SNAr) to synthesize hydrogels under mild conditions in a buffer solution, the necessity of additives or posttreatments can be obviated. The nucleophilic and redox reactions between selenol and tetrazine culminate in the formation of three degradable chemical bonds-diselenide, aryl selenide, and dearomatized selenide-in a single, expeditious step. The resultant hydrogel manifests exceptional adaptability to intricate environments in conjunction with self-healing and on-demand degradation properties. Furthermore, the resulting material demonstrated light-triggered antibacterial activity. Animal studies further underscore the potential of integrating metformin into Se-Tz hydrogels under green light irradiation, as it effectively stimulates angiogenesis and collagen deposition, thereby fostering efficient wound healing. In comparison to previously documented hydrogels, Se-Tz hydrogels exhibit controlled degradation and drug release, outstanding antibacterial activity, mechanical robustness, and bioactivity, all without the need for costly and intricate preparation procedures. These findings underscore Se-Tz hydrogels as a safe and effective therapeutic option for diabetic wound dressings.
Collapse
Affiliation(s)
- Yan Wu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Ying Bei
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
- Hainan Academy of Medical Sciences, Hainan Medical University, Hainan, 571199, China
| | - Wenjing Li
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Weihong Lu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Jian Zhu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Zhengbiao Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Sen Liu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Kaiyuan Chen
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Hong Jin
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Luxin Li
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Meng Li
- Department of Dermatology Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200010, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Xiangqiang Pan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| |
Collapse
|
14
|
Mishra A, Kushare A, Gupta MN, Ambre P. Advanced Dressings for Chronic Wound Management. ACS APPLIED BIO MATERIALS 2024; 7:2660-2676. [PMID: 38723276 DOI: 10.1021/acsabm.4c00138] [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: 05/21/2024]
Abstract
Wound healing, particularly for chronic wounds, presents a considerable difficulty due to differences in biochemical and cellular processes that occur in different types of wounds. Recent technological breakthroughs have notably advanced the understanding of diagnostic and therapeutic approaches to wound healing. The evolution in wound care has seen a transition from traditional textile dressings to a variety of advanced alternatives, including self-healing hydrogels, hydrofibers, foams, hydrocolloids, environment responsive dressings, growth factor-based therapy, bioengineered skin substitutes, and stem cell and gene therapy. Technological advancements, such as 3D printing and electronic skin (e-skin) therapy, contribute to the customization of wound healing. Despite these advancements, effectively managing chronic wounds remains challenging. This necessitates the development of treatments that consider performance, risk-benefit balance, and cost-effectiveness. This review discusses innovative strategies for the healing of chronic wounds. Incorporating biomarkers into advanced dressings, coupled with corresponding biosensors and drug delivery formulations, enables the theranostic approach to the treatment of chronic wounds. Furthermore, integrating advanced dressings with power sources and user interfaces like near-field communication, radio frequency identification, and Bluetooth enhances real-time monitoring and on-demand drug delivery. It also provides a thorough evaluation of the advantages, patient compliance, costs, and durability of advanced dressings, emphasizing smart formulations and their preparation methods.
Collapse
Affiliation(s)
- Abhishek Mishra
- Department of Pharmaceutical Chemistry, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai 400098, India
| | - Aniket Kushare
- Department of Pharmaceutical Chemistry, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai 400098, India
| | - Munishwar Nath Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Premlata Ambre
- Department of Pharmaceutical Chemistry, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai 400098, India
| |
Collapse
|
15
|
Wang Y, He X, Huang K, Cheng N. Nanozyme as a rising star for metabolic disease management. J Nanobiotechnology 2024; 22:226. [PMID: 38711066 PMCID: PMC11071342 DOI: 10.1186/s12951-024-02478-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024] Open
Abstract
Nanozyme, characterized by outstanding and inherent enzyme-mimicking properties, have emerged as highly promising alternatives to natural enzymes owning to their exceptional attributes such as regulation of oxidative stress, convenient storage, adjustable catalytic activities, remarkable stability, and effortless scalability for large-scale production. Given the potent regulatory function of nanozymes on oxidative stress and coupled with the fact that reactive oxygen species (ROS) play a vital role in the occurrence and exacerbation of metabolic diseases, nanozyme offer a unique perspective for therapy through multifunctional activities, achieving essential results in the treatment of metabolic diseases by directly scavenging excess ROS or regulating pathologically related molecules. The rational design strategies, nanozyme-enabled therapeutic mechanisms at the cellular level, and the therapies of nanozyme for several typical metabolic diseases and underlying mechanisms are discussed, mainly including obesity, diabetes, cardiovascular disease, diabetic wound healing, and others. Finally, the pharmacokinetics, safety analysis, challenges, and outlooks for the application of nanozyme are also presented. This review will provide some instructive perspectives on nanozyme and promote the development of enzyme-mimicking strategies in metabolic disease therapy.
Collapse
Affiliation(s)
- Yanan Wang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing, 100083, People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China
| | - Xiaoyun He
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing, 100083, People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China
| | - Kunlun Huang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing, 100083, People's Republic of China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China.
| | - Nan Cheng
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing, 100083, People's Republic of China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China.
| |
Collapse
|
16
|
Wu Y, Li L, Ning Z, Li C, Yin Y, Chen K, Li L, Xu F, Gao J. Autophagy-modulating biomaterials: multifunctional weapons to promote tissue regeneration. Cell Commun Signal 2024; 22:124. [PMID: 38360732 PMCID: PMC10868121 DOI: 10.1186/s12964-023-01346-3] [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: 08/24/2023] [Accepted: 09/29/2023] [Indexed: 02/17/2024] Open
Abstract
Autophagy is a self-renewal mechanism that maintains homeostasis and can promote tissue regeneration by regulating inflammation, reducing oxidative stress and promoting cell differentiation. The interaction between biomaterials and tissue cells significantly affects biomaterial-tissue integration and tissue regeneration. In recent years, it has been found that biomaterials can affect various processes related to tissue regeneration by regulating autophagy. The utilization of biomaterials in a controlled environment has become a prominent approach for enhancing the tissue regeneration capabilities. This involves the regulation of autophagy in diverse cell types implicated in tissue regeneration, encompassing the modulation of inflammatory responses, oxidative stress, cell differentiation, proliferation, migration, apoptosis, and extracellular matrix formation. In addition, biomaterials possess the potential to serve as carriers for drug delivery, enabling the regulation of autophagy by either activating or inhibiting its processes. This review summarizes the relationship between autophagy and tissue regeneration and discusses the role of biomaterial-based autophagy in tissue regeneration. In addition, recent advanced technologies used to design autophagy-modulating biomaterials are summarized, and rational design of biomaterials for providing controlled autophagy regulation via modification of the chemistry and surface of biomaterials and incorporation of cells and molecules is discussed. A better understanding of biomaterial-based autophagy and tissue regeneration, as well as the underlying molecular mechanisms, may lead to new possibilities for promoting tissue regeneration. Video Abstract.
Collapse
Affiliation(s)
- Yan Wu
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Luxin Li
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Zuojun Ning
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Changrong Li
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Yongkui Yin
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Kaiyuan Chen
- Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Lu Li
- Department of plastic surgery, Naval Specialty Medical Center of PLA, Shanghai, 200052, China.
| | - Fei Xu
- Department of plastic surgery, Naval Specialty Medical Center of PLA, Shanghai, 200052, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
17
|
Morrow JP, Mazrad ZAI, Warne NM, Ayton S, Bush AI, Kempe K. Schiff-Base Cross-Linked Poly(2-oxazoline) Micelle Drug Conjugates Possess Antiferroptosis Activity in Numerous In Vitro Cell Models. Biomacromolecules 2024; 25:1068-1083. [PMID: 38178625 DOI: 10.1021/acs.biomac.3c01106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
A great deal of nanocarriers have been applied to induce ferroptosis in cancer research, yet there are limited examples of nanocarrier formulations to rescue ferroptosis, which can be applied to neurodegeneration, inflammation, liver damage, kidney disease, and more. Here, we present the synthesis, characterization, and in vitro evaluation of pH-responsive, core-cross-linked micelle (CCM) ferrostatin-1 (Fer-1) conjugates with amine, valproic acid, and biotin surface chemistries. Fer-1 release from stable and defined CCM Fer-1 conjugates was quantified, highlighting the sustained release for 24 h. CCM Fer-1 conjugates demonstrated excellent ferroptosis rescue by their antilipid peroxidation activity in a diverse set of cell lines in vitro. Additionally, CCMs showed tunable cell association in SH-SY5Y and translocation across an in vitro blood-brain barrier (BBB) model, highlighting potential brain disease applications. Overall, here, we present a polymeric Fer-1 delivery system to enhance Fer-1 action, which could help in improving Fer-1 action in the treatment of ferroptosis-related diseases.
Collapse
Affiliation(s)
- Joshua P Morrow
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Zihnil A I Mazrad
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Nicole M Warne
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
18
|
Kurian AG, Singh RK, Sagar V, Lee JH, Kim HW. Nanozyme-Engineered Hydrogels for Anti-Inflammation and Skin Regeneration. NANO-MICRO LETTERS 2024; 16:110. [PMID: 38321242 PMCID: PMC10847086 DOI: 10.1007/s40820-024-01323-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 12/24/2023] [Indexed: 02/08/2024]
Abstract
Inflammatory skin disorders can cause chronic scarring and functional impairments, posing a significant burden on patients and the healthcare system. Conventional therapies, such as corticosteroids and nonsteroidal anti-inflammatory drugs, are limited in efficacy and associated with adverse effects. Recently, nanozyme (NZ)-based hydrogels have shown great promise in addressing these challenges. NZ-based hydrogels possess unique therapeutic abilities by combining the therapeutic benefits of redox nanomaterials with enzymatic activity and the water-retaining capacity of hydrogels. The multifaceted therapeutic effects of these hydrogels include scavenging reactive oxygen species and other inflammatory mediators modulating immune responses toward a pro-regenerative environment and enhancing regenerative potential by triggering cell migration and differentiation. This review highlights the current state of the art in NZ-engineered hydrogels (NZ@hydrogels) for anti-inflammatory and skin regeneration applications. It also discusses the underlying chemo-mechano-biological mechanisms behind their effectiveness. Additionally, the challenges and future directions in this ground, particularly their clinical translation, are addressed. The insights provided in this review can aid in the design and engineering of novel NZ-based hydrogels, offering new possibilities for targeted and personalized skin-care therapies.
Collapse
Affiliation(s)
- Amal George Kurian
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Rajendra K Singh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Varsha Sagar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Cell and Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea.
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea.
- Cell and Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea.
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
19
|
Wang X, Yang Y, Zhao W, Zhu Z, Pei X. Recent advances of hydrogels as smart dressings for diabetic wounds. J Mater Chem B 2024; 12:1126-1148. [PMID: 38205636 DOI: 10.1039/d3tb02355a] [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: 01/12/2024]
Abstract
Chronic diabetic wounds have been an urgent clinical problem, and wound dressings play an important role in their management. Due to the design of traditional dressings, it is difficult to achieve adaptive adhesion and on-demand removal of complex diabetic wounds, real-time monitoring of wound status, and dynamic adjustment of drug release behavior according to the wound microenvironment. Smart hydrogels, as smart dressings, can respond to environmental stimuli and achieve more precise local treatment. Here, we review the latest progress of smart hydrogels in wound bandaging, dynamic monitoring, and drug delivery for treatment of diabetic wounds. It is worth noting that we have summarized the most important properties of smart hydrogels for diabetic wound healing. In addition, we discuss the unresolved challenges and future prospects in this field. We hope that this review will contribute to furthering progress on smart hydrogels as improved dressing for diabetic wound healing and practical clinical application.
Collapse
Affiliation(s)
- Xu Wang
- 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, No. 14, Section 3, South Peoples Road, Chengdu, 610041, Sichuan, China.
| | - Yuhan Yang
- 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, No. 14, Section 3, South Peoples Road, Chengdu, 610041, Sichuan, China.
| | - Weifeng Zhao
- College of Polymer Science and Engineering, The State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Zhou Zhu
- 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, No. 14, Section 3, South Peoples Road, Chengdu, 610041, Sichuan, China.
| | - Xibo Pei
- 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, No. 14, Section 3, South Peoples Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
20
|
Wang Y, Zhang M, Yan Z, Ji S, Xiao S, Gao J. Metal nanoparticle hybrid hydrogels: the state-of-the-art of combining hard and soft materials to promote wound healing. Theranostics 2024; 14:1534-1560. [PMID: 38389845 PMCID: PMC10879867 DOI: 10.7150/thno.91829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/27/2023] [Indexed: 02/24/2024] Open
Abstract
Wounds represent a grave affliction that profoundly impacts human well-being. Establishing barriers, preventing infections, and providing a conducive microenvironment constitute the crux of wound therapy. Hydrogel, a polymer with an intricate three-dimensional lattice, serves as a potent tool in erecting physical barriers and nurturing an environment conducive to wound healing. This enables effective control over exudation, hemostasis, accelerated wound closure, and diminished scar formation. As a result, hydrogels have gained extensive traction in the realm of wound treatment. Metallic nanoparticle carriers, characterized by their multifaceted responses encompassing acoustics, optics, and electronics, have demonstrated efficacy in wound management. Nevertheless, these carriers encounter challenges associated with swift clearance and nonuniform effectiveness. The hybridization of metallic nanoparticle carriers with hydrogels overcomes the shortcomings inherent in metallic nanoparticle-based wound therapy. This amalgamation not only addresses the limitations but also augments the mechanical robustness of hydrogels. It confers upon them attributes such as environmental responsiveness and multifunctionality, thereby synergizing strengths and compensating for weaknesses. This integration culminates in the precise and intelligent management of wounds. This review encapsulates the structural classifications, design strategies, therapeutic applications, and underlying mechanisms of metal nanoparticle hybrid hydrogels in the context of acute and chronic wound treatment. The discourse delves into the generation of novel or enhanced attributes arising from hybridization and how the current paradigm of wound therapy leverages these attributes. Amidst this continually evolving frontier, the potential of metal nanoparticle hybrid hydrogels to revolutionize wound treatment is underscored.
Collapse
Affiliation(s)
- Yuxiang Wang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Mengya Zhang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Zhenzhen Yan
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Jie Gao
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, People's Republic of China
| |
Collapse
|
21
|
Zhou Z, Liu Y, Li W, Zhao Z, Xia X, Liu J, Deng Y, Wu Y, Pan X, He F, Yang H, Lu W, Xu Y, Zhu X. A Self-Adaptive Biomimetic Periosteum Employing Nitric Oxide Release for Augmenting Angiogenesis in Bone Defect Regeneration. Adv Healthc Mater 2024; 13:e2302153. [PMID: 37922941 DOI: 10.1002/adhm.202302153] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/12/2023] [Indexed: 11/07/2023]
Abstract
The periosteum plays a vital role in the regeneration of critical-size bone defects and highly comminuted fractures, promoting the differentiation of osteoblasts, accelerating the reconstruction of the vascular network, and guiding bone tissue regeneration. However, the materials loaded with exogenous growth factors are limited by the release and activity of the elements. Therefore, the material structure must be carefully designed for the periosteal function. Here, a self-adaptive biomimetic periosteum strategy is proposed, which is a novel interpenetrating double network hydrogel consisting of diselenide-containing gelatin and calcium alginate (modified natural collagen and polysaccharide) to enhance the stability, anti-swelling, and delayed degradation of the hydrogel. The diselenide bond continuously releases nitric oxide (NO) by metabolizing endogenous nitrosated thiols (RSNO), activates the nitric oxide-cycle guanosine monophosphate (NO-cGMP) signal pathway, coordinates the coupling effect of angiogenesis and osteogenesis, and accelerates the repair of bone defects. This self-adaptive biomimetic periosteum with the interpenetrating double network structure formed by the diselenide-containing gelatin and calcium alginate has been proven to be safe and effective in repairing critical-size bone defects and is expected to provide a promising strategy for solving clinical problems.
Collapse
Affiliation(s)
- Zhangzhe Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Yang Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Wenjing Li
- Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Zhijian Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Xiaowei Xia
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Junlin Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Yaoge Deng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Yubin Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Xiangqiang Pan
- Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Weihong Lu
- Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Yong Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| |
Collapse
|
22
|
Jeyachandran S, Srinivasan R, Ramesh T, Parivallal A, Lee J, Sathiyamoorthi E. Recent Development and Application of "Nanozyme" Artificial Enzymes-A Review. Biomimetics (Basel) 2023; 8:446. [PMID: 37754197 PMCID: PMC10526256 DOI: 10.3390/biomimetics8050446] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Nanozymes represent a category of nano-biomaterial artificial enzymes distinguished by their remarkable catalytic potency, stability, cost-effectiveness, biocompatibility, and degradability. These attributes position them as premier biomaterials with extensive applicability across medical, industrial, technological, and biological domains. Following the discovery of ferromagnetic nanoparticles with peroxidase-mimicking capabilities, extensive research endeavors have been dedicated to advancing nanozyme utilization. Their capacity to emulate the functions of natural enzymes has captivated researchers, prompting in-depth investigations into their attributes and potential applications. This exploration has yielded insights and innovations in various areas, including detection mechanisms, biosensing techniques, and device development. Nanozymes exhibit diverse compositions, sizes, and forms, resembling molecular entities such as proteins and tissue-based glucose. Their rapid impact on the body necessitates a comprehensive understanding of their intricate interplay. As each day witnesses the emergence of novel methodologies and technologies, the integration of nanozymes continues to surge, promising enhanced comprehension in the times ahead. This review centers on the expansive deployment and advancement of nanozyme materials, encompassing biomedical, biotechnological, and environmental contexts.
Collapse
Affiliation(s)
- Sivakamavalli Jeyachandran
- Laboratory in Biotechnology & Biosignal Transduction, Department of Orthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Ramachandran Srinivasan
- Centre for Ocean Research (DST-FIST Sponsored Centre), MoES-Earth Science and Technology Cell (Marine Biotechnological Studies), Sathyabama Research Park, Sathyabama Institute of Science and Technology, Chennai 600119, Tamil Nadu, India;
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia;
| | - Arumugam Parivallal
- Department of Mathematics, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | | |
Collapse
|
23
|
Jiang X, Wu L, Zhang M, Zhang T, Chen C, Wu Y, Yin C, Gao J. Biomembrane nanostructures: Multifunctional platform to enhance tumor chemoimmunotherapy via effective drug delivery. J Control Release 2023; 361:510-533. [PMID: 37567505 DOI: 10.1016/j.jconrel.2023.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/02/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Chemotherapeutic drugs have been found to activate the immune response against tumors by inducing immunogenic cell death, in addition to their direct cytotoxic effects toward tumors, therefore broadening the application of chemotherapy in tumor immunotherapy. The combination of other therapeutic strategies, such as phototherapy or radiotherapy, could further strengthen the therapeutic effects of immunotherapy. Nanostructures can facilitate multimodal tumor therapy by integrating various active agents and combining multiple types of therapeutics in a single nanostructure. Biomembrane nanostructures (e.g., exosomes and cell membrane-derived nanostructures), characterized by superior biocompatibility, intrinsic targeting ability, intelligent responsiveness and immune-modulating properties, could realize superior chemoimmunotherapy and represent next-generation nanostructures for tumor immunotherapy. This review summarizes recent advances in biomembrane nanostructures in tumor chemoimmunotherapy and highlights different types of engineering approaches and therapeutic mechanisms. A series of engineering strategies for combining different biomembrane nanostructures, including liposomes, exosomes, cell membranes and bacterial membranes, are summarized. The combination strategy can greatly enhance the targeting, intelligence and functionality of biomembrane nanostructures for chemoimmunotherapy, thereby serving as a stronger tumor therapeutic method. The challenges associated with the clinical translation of biomembrane nanostructures for chemoimmunotherapy and their future perspectives are also discussed.
Collapse
Affiliation(s)
- Xianghe Jiang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; College of Life Science, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Lili Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Mengya Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Cuimin Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang 157011, China.
| | - Chuan Yin
- Department of Gastroenterology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|