1
|
Xiao J, Song Z, Liu T, Guo Z, Liu X, Jiang H, Wang X. Cell Membrane Engineered Polypeptide Nanonets Mimicking Macrophage Aggregates for Enhanced Antibacterial Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401845. [PMID: 38966869 DOI: 10.1002/smll.202401845] [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: 03/07/2024] [Revised: 06/18/2024] [Indexed: 07/06/2024]
Abstract
Drug-resistant bacterial infections and their lipopolysaccharide-related inflammatory complications continue to pose significant challenges in traditional treatments. Inspired by the rapid initiation of resident macrophages to form aggregates for efficient antibacterial action, this study proposes a multifunctional and enhanced antibacterial strategy through the construction of novel biomimetic cell membrane polypeptide nanonets (R-DPB-TA-Ce). The design involves the fusion of end-terminal lipidated polypeptides containing side-chain cationic boronic acid groups (DNPLBA) with cell membrane intercalation engineering (R-DPB), followed by coordination with the tannic acid-cerium complex (TA-Ce) to assemble into a biomimetic nanonet through boronic acid-polyphenol-metal ion interactions. In addition to the ability of RAW 264.7 macrophages cell membrane components' (R) ability to neutralize lipopolysaccharide (LPS), R-DPB-TA-Ce demonstrated enhanced capture of bacteria and its LPS, leveraging nanoconfinement-enhanced multiple interactions based on the boronic acid-polyphenol nanonets skeleton combined with polysaccharide. Utilizing these advantages, indocyanine green (ICG) is further employed as a model drug for delivery, showcasing the exceptional treatment effect of R-DPB-TA-Ce as a new biomimetic assembled drug delivery system in antibacterial, anti-inflammatory, and wound healing promotion. Thus, this strategy of mimicking macrophage aggregates is anticipated to be further applicable to various types of cell membrane engineering for enhanced antibacterial treatment.
Collapse
Affiliation(s)
- Jiang Xiao
- Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Zhongquan Song
- Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Tengfei Liu
- Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Zengchao Guo
- Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Xiaohui Liu
- Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Hui Jiang
- Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Xuemei Wang
- Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, China
| |
Collapse
|
2
|
Mo XW, Phan NM, Nguyen TL, Kim J. H 2O 2 Self-Supplying CaO 2 Nanoplatform Induces Ca 2+ Overload Combined with Chemodynamic Therapy to Enhance Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58337-58345. [PMID: 39406705 DOI: 10.1021/acsami.4c12748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Integrating chemodynamic therapy (CDT) with Ca2+ overload offers a potent strategy for enhancing cancer immunotherapy. However, the effectiveness of this approach is significantly constrained by the scarce availability of H2O2 in solid tumors. Here, we engineered a nanoplatform based on CaO2 nanoparticles (NPs) capable of encapsulating curcumin (CUR) and self-supplying H2O2 for synergistic CDT-augmented antitumor immunotherapy (CaO2@CUR@ZIF-Cu, denoted as CCZC). In the acidic tumor microenvironment, CCZC disintegrated to release CUR and copper(II) ions (Cu2+), revealing the core CaO2 NPs. CDT was amplified by escalating hydroxyl radical (•OH) production through a Fenton-like reaction mediated by H2O2 from the hydrolysis of CaO2 NPs. Ca2+ sourced from CaO2 NPs and CUR, an initiator of Ca2+ overload, induced Ca2+ overload in tumor cells, thereby promoting apoptosis. Subsequently, apoptotic tumor cells released tumor-associated antigens and pro-inflammatory cytokines, triggering adaptive immune responses and enhancing antitumor immunotherapy effects. In vivo experiments demonstrated that the intratumoral administration of CCZC displayed significant inhibitory effects, with an inhibition rate of up to 78% on B16-OVA-tumor-bearing mice compared to untreated. Moreover, an elevated proportion of mature dendritic cells were observed in the tumor-draining lymph nodes, along with an increase in cytotoxic T lymphocytes in the spleen. These findings suggest that our engineered nanoplatform effectively curtailed tumor growth via enhanced cancer immunotherapy by synergizing Ca2+ overload and CDT, proposing a novel strategy for synergistic cancer treatment.
Collapse
Affiliation(s)
- Xin Wang Mo
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ngoc Man Phan
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Thanh Loc Nguyen
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- South Australian ImmunoGENomics Cancer Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, South Australia, Australia
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of MetaBioHealth, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
3
|
Tagaras N, Song H, Sahar S, Tong W, Mao Z, Buerki-Thurnherr T. Safety Landscape of Therapeutic Nanozymes and Future Research Directions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2407816. [PMID: 39445544 DOI: 10.1002/advs.202407816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/20/2024] [Indexed: 10/25/2024]
Abstract
Oxidative stress and inflammation are at the root of a multitude of diseases. Treatment of these conditions is often necessary but current standard therapies to fight excessive reactive oxygen species (ROS) and inflammation are often ineffective or complicated by substantial safety concerns. Nanozymes are emerging nanomaterials with intrinsic enzyme-like properties that hold great promise for effective cancer treatment, bacterial elimination, and anti-inflammatory/anti-oxidant therapy. While there is rapid progress in tailoring their catalytic activities as evidenced by the recent integration of single-atom catalysts (SACs) to create next-generation nanozymes with superior activity, selectivity, and stability, a better understanding and tuning of their safety profile is imperative for successful clinical translation. This review outlines the current applied safety assessment approaches and provides a comprehensive summary of the safety knowledge of therapeutic nanozymes. Overall, nanozymes so far show good in vitro and in vivo biocompatibility despite considerable differences in their composition and enzymatic activities. However, current safety investigations mostly cover a limited set of basic toxicological endpoints, which do not allow for a thorough and deep assessment. Ultimately, remaining research gaps that should be carefully addressed in future studies are highlighted, to optimize the safety profile of therapeutic nanozymes early in their pre-clinical development.
Collapse
Affiliation(s)
- Nikolaos Tagaras
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen, 9014, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, 8093, Switzerland
| | - Haihan Song
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Shafaq Sahar
- College of Chemical and Biological Engineering, MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Weijun Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Zhengwei Mao
- College of Chemical and Biological Engineering, MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Tina Buerki-Thurnherr
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen, 9014, Switzerland
| |
Collapse
|
4
|
Zhao J, Wang Y, Zhang Y, Guo X, Bao H, Tao Y. In Situ Crystallized Ceria-Vesicle Nanohybrid Therapeutic for Effective Treatment of Inflammatory Intraocular Disease. Adv Healthc Mater 2024:e2402523. [PMID: 39440628 DOI: 10.1002/adhm.202402523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/21/2024] [Indexed: 10/25/2024]
Abstract
Posterior uveitis is a leading cause of vision impairment and blindness globally due to its detrimental effects on the choroid and retina. The condition is worsened by oxidative stress, which heightens inflammation and perpetuates a cycle of damage that current treatments only temporarily relieve. To address this, a novel treatment involving the in situ crystallization of ultrasmall cerium oxide nanoparticles (≈3 nm) on mesenchymal stem cell (MSC) extracellular vesicles (EVs) for the management of primed mycobacterial uveitis (PMU) is developed. This nanohybrid leverages the individual and synergistic effects of its components for a comprehensive therapeutic approach. The cerium oxide nanoparticles act as a nanozyme to reduce inflammation and scavenge excessive reactive oxygen species (ROS), while the MSC EVs, with their biocompatibility, modulate inflammatory cell infiltration and alleviate tissue damage. This synergistic system offers a promising new treatment strategy for ocular diseases characterized by oxidative stress and inflammation.
Collapse
Affiliation(s)
- Jiawei Zhao
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, P. R. China
| | - Yingjie Wang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, P. R. China
| | - Yiquan Zhang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, P. R. China
| | - Xinyu Guo
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, P. R. China
| | - Han Bao
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, P. R. China
| | - Yong Tao
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, P. R. China
| |
Collapse
|
5
|
Zhang Y, Wu D, Zhou C, Bai M, Wan Y, Zheng Q, Fan Z, Wang X, Yang C. Engineered extracellular vesicles for tissue repair and regeneration. BURNS & TRAUMA 2024; 12:tkae062. [PMID: 39439545 PMCID: PMC11495891 DOI: 10.1093/burnst/tkae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) are heterogeneous membrane-like vesicles secreted by living cells that are involved in many physiological and pathological processes and act as intermediaries of intercellular communication and molecular transfer. Recent studies have shown that EVs from specific sources regulate tissue repair and regeneration by delivering proteins, lipids, and nucleic acids to target cells as signaling molecules. Nanotechnology breakthroughs have facilitated the development and exploration of engineered EVs for tissue repair. Enhancements through gene editing, surface modification, and content modification have further improved their therapeutic efficacy. This review summarizes the potential of EVs in tissue repair and regeneration, their mechanisms of action, and their research progress in regenerative medicine. This review highlights their design logic through typical examples and explores the development prospects of EVs in tissue repair. The aim of this review is to provide new insights into the design of EVs for tissue repair and regeneration applications, thereby expanding their use in regenerative medicine.
Collapse
Affiliation(s)
- Yan Zhang
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
- School of Public Health, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Dan Wu
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Chen Zhou
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-Sen University, No. 3025 Shennan Middle Road, Futian District, Shenzhen, China
| | - Muran Bai
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Yucheng Wan
- Hospital of Stomatology, Zunyi Medical University, No. 89, Wujiang East Road, Xinpu New District, Zunyi City, Guizhou Province, China
| | - Qing Zheng
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Zhijin Fan
- Institute for Engineering Medicine, Kunming Medical University, No. 1168 Chunrong West Road, Yuhua Street, Chenggong District, Kunming City, Yunnan Province China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, No.81 Meishan Road, Shushan District, Hefei 230032, China
| | - Chun Yang
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| |
Collapse
|
6
|
Jiang X, Wei J, Ding X, Zheng K, Zhou T, Shi J, Lai H, Qian S, Zhang X. From ROS scavenging to boosted osseointegration: cerium-containing mesoporous bioactive glass nanoparticles functionalized implants in diabetes. J Nanobiotechnology 2024; 22:639. [PMID: 39425200 PMCID: PMC11488221 DOI: 10.1186/s12951-024-02865-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: 01/19/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024] Open
Abstract
Excessive production of reactive oxygen species (ROS) around titanium implants under diabetic conditions causes persistent inflammation, leading to poor osseointegration and even implant failure. Surface modification is an effective way to promote ROS clearance, alleviate inflammation, and stimulate bone formation. In this study, a multifunctional coating is fabricated by introducing cerium (Ce)-containing mesoporous bioactive glass nanoparticles (Ce-MBGNs) onto the titanium surface via an electrophoretic deposition method. The incorporation of Ce-MBGNs remarkably improves surface hydrophilicity by increasing the surface areas. The bioactive ions are appropriately released, thereby promoting mesenchymal stem cell proliferation and differentiation under diabetic conditions. The conversion between Ce(III) and Ce(IV) endows Ce-MBGNs coating with antioxidative nanoenzymes properties to scavenge diabetes-induced ROS, resulting in macrophage polarization towards the anti-inflammatory phenotype. The therapeutic effect of Ce-MBGNs-modified titanium implants is also verified in diabetic rats by inhibiting inflammatory responses and accelerating early osseointegration. Taken together, the findings reveal that the ROS-scavenging and immunomodulation activity of the Ce-MBGNs coating contributes to enhanced osseointegration, and provides a novel implant surface for diabetic patients.
Collapse
Affiliation(s)
- Xue Jiang
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; , Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Jianxu Wei
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; , Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xinxin Ding
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; , Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Kai Zheng
- Jiangsu Key Laboratory of Oral Diseases and Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Tian Zhou
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; , Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Junyu Shi
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; , Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Hongchang Lai
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; , Shanghai Research Institute of Stomatology, Shanghai, 200011, China.
| | - Shujiao Qian
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; , Shanghai Research Institute of Stomatology, Shanghai, 200011, China.
| | - Xiaomeng Zhang
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; , Shanghai Research Institute of Stomatology, Shanghai, 200011, China.
| |
Collapse
|
7
|
Song P, Wu Y, Fan M, Chen X, Dong M, Qiao W, Dong N, Wang Q. Folic acid modified silver nanoparticles promote endothelialization and inhibit calcification of decellularized heart valves by immunomodulation with anti-bacteria property. BIOMATERIALS ADVANCES 2024; 166:214069. [PMID: 39447240 DOI: 10.1016/j.bioadv.2024.214069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024]
Abstract
Xenogeneic decellularized heart valves (DHVs) have become one of the most commonly used scaffolds for tissue engineered heart valves (TEHVs) due to extensive resources and possessing the distinct three-layer structure similar to native heart valves. However, DHVs as scaffolds face the shortages such as poor mechanical properties, proneness to thrombosis and calcification, difficulty in endothelialization and chronic inflammatory responses etc., which limit their applications in clinic. In this work, we constructed a novel TEHV with immunomodulatory functions by loading folic acid modified silver nanoparticles (FS NPs) on DHVs to overcome these issues. The FS NPs preferentially targeted M1 macrophages and reduced their intracellular H2O2 level, resulting in polarizing them into M2 phenotype. The increased M2 macrophages facilitated to eliminate inflammation, recruit endothelial cells, and promote their proliferation and endothelialization by secreting relative factors. We founded that FS NPs with the size of 80 nm modified DHVs (FSD-80) performed optimally on cytocompatibility and regulating macrophage phenotype ability in vitro. In addition, the FSD-80 had excellent mechanical properties, hemocompatibility and anti-bacteria property. The results of the subcutaneous implantation in rats revealed that the FSD-80 also had good performance in regulating macrophage phenotype, promoting endothelialization, remolding the extracellular matrix and anti-calcification in vivo. Therefore, FS NPs-loaded DHVs possess immunomodulatory functions, which is a feasible and promising strategy for constructing TEHVs with excellent comprehensive performance.
Collapse
Affiliation(s)
- Peng Song
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Centre for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yunlong Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Min Fan
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Centre for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Cardiovascular Surgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Mengna Dong
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Centre for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Qin Wang
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Centre for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
8
|
Wang Y, Liao Y, Zhang YJ, Wu XH, Qiao ZY, Wang H. Self-Assembled Peptide with Morphological Structure for Bioapplication. Biomacromolecules 2024; 25:6367-6394. [PMID: 39297513 DOI: 10.1021/acs.biomac.4c01179] [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: 10/15/2024]
Abstract
Peptide materials, such as self-assembled peptide materials, are very important biomaterials. Driven by multiple interaction forces, peptide molecules can self-assemble into a variety of different macroscopic forms with different properties and functions. In recent years, the research on self-assembled peptides has made great progress from laboratory design to clinical application. This review focuses on the different morphologies, including nanoparticles, nanovesicles, nanotubes, nanofibers, and others, formed by self-assembled peptide. The mechanisms and applications of the morphology transformation are also discussed in this paper, and the future direction of self-assembled nanomaterials is envisioned.
Collapse
Affiliation(s)
- Yu Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Yusi Liao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, P. R. China
| | - Ying-Jin Zhang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Xiu-Hai Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
- Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin150081, P. R. China
| | - Zeng-Ying Qiao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Hao Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, P. R. China
| |
Collapse
|
9
|
Tian S, Mei J, Zhang L, Wang S, Yuan Y, Li J, Liu H, Zhu W, Xu D. Multifunctional Hydrogel Microneedle Patches Modulating Oxi-inflamm-aging for Diabetic Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2407340. [PMID: 39360460 DOI: 10.1002/smll.202407340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/21/2024] [Indexed: 10/04/2024]
Abstract
Oxidative stress, chronic inflammation, and immune senescence are important pathologic factors in diabetic wound nonhealing. This study loads taurine (Tau) into cerium dioxide (CeO2) to develop CeO2@Tau nanoparticles with excellent antioxidant, anti-inflammatory, and anti-aging properties. To enhance the drug penetration efficiency in wounds, CeO2@Tau is encapsulated in gelatin methacryloyl (GelMA) hydrogel to prepare CeO2@Tau@Hydrogel@Microneedle (CTH@MN) patch system. Microneedle technology achieves precise and efficient delivery of CeO2@Tau, ensuring their deep penetration into the wound tissue for optimal efficacy. Rigorous in vitro and in vivo tests have confirmed the satisfactory therapeutic effect of CTH@MN patch on diabetic wound healing. Mechanistically, CTH@MN attenuates oxidative damage and inflammatory responses in macrophages by inhibiting the ROS/NF-κB signaling pathway. Meanwhile, CTH@MN activated autophagy-mediated anti-aging activity, creating a favorable immune microenvironment for tissue repair. Notably, in a diabetic mouse wound model, the multifunctional CTH@MN patch significantly promotes wound healing by systematically regulating the oxidation-inflammation-aging (oxi-inflamm-aging) pathological axis. In conclusion, the in-depth exploration of the CTH@MN system in this study provides new strategies and perspectives for treating diabetic non-healing wounds.
Collapse
Affiliation(s)
- Shen Tian
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Jiawei Mei
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, P. R. China
| | - Lisha Zhang
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Senyan Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Yuhui Yuan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Jia Li
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Wanbo Zhu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, P. R. China
| | - Dongdong Xu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| |
Collapse
|
10
|
Cui M, Qian L, Lu K, Liu J, Chu B, Wu X, Dong F, Song B, He Y. Defect-Rich Metastable MoS 2 Promotes Macrophage Reprogramming in Breast Cancer: A Clinical Perspective. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402101. [PMID: 38888117 DOI: 10.1002/smll.202402101] [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: 03/17/2024] [Revised: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Tumor-associated macrophages (TAMs) play a crucial function in solid tumor antigen clearance and immune suppression. Notably, 2D transitional metal dichalcogenides (i.e., molybdenum disulfide (MoS2) nanozymes) with enzyme-like activity are demonstrated in animal models for cancer immunotherapy. However, in situ engineering of TAMs polarization through sufficient accumulation of free radical reactive oxygen species for immunotherapy in clinical samples remains a significant challenge. In this study, defect-rich metastable MoS2 nanozymes, i.e., 1T2H-MoS2, are designed via reduction and phase transformation in molten sodium as a guided treatment for human breast cancer. The as-prepared 1T2H-MoS2 exhibited enhanced peroxidase-like activity (≈12-fold enhancement) than that of commercial MoS2, which is attributed to the charge redistribution and electronic state induced by the abundance of S vacancies. The 1T2H-MoS2 nanozyme can function as an extracellular hydroxyl radical generator, efficiently repolarizing TAMs into the M1-like phenotype and directly killing cancer cells. Moreover, the clinical feasibility of 1T2H-MoS2 is demonstrated via ex vivo therapeutic responses in human breast cancer samples. The apoptosis rate of cancer cells is 3.4 times greater than that of cells treated with chemotherapeutic drugs (i.e., doxorubicin).
Collapse
Affiliation(s)
- Mingyue Cui
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Lulu Qian
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Ke Lu
- Institutes of Physical Science and Information Technology, Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Anhui University, Hefei, Anhui, 230601, China
| | - Jinjin Liu
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Binbin Chu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Xiaofeng Wu
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Fenglin Dong
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Bin Song
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Yao He
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Macao Translational Medicine Center, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| |
Collapse
|
11
|
Baik S, Kim H, Lee Y, Kang T, Shin K, Song C, Park OK, Kang B, Lee N, Kim D, Choi SH, Kim SH, Soh M, Hyeon T, Kim CK. Orally Deliverable Iron-Ceria Nanotablets for Treatment of Inflammatory Bowel Disease. Adv Healthc Mater 2024:e2401994. [PMID: 39235381 DOI: 10.1002/adhm.202401994] [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: 08/20/2024] [Indexed: 09/06/2024]
Abstract
Ceria-based nanoparticles are versatile in treating various inflammatory diseases, but their feasibility in clinical translation is undermined by safety concerns and a limited delivery system. Meanwhile, the idiopathic nature of inflammatory bowel disease (IBD) calls for a wider variety of therapeutics via moderation of the intestinal immune system. In this regard, the synthesis and oral formulation of iron-ceria nanoparticles (CF NPs) with enhanced nanozymic activity and lower toxicity risk than conventional ceria-based nanoparticles are reported. CF NPs are clustered in calcium phosphate (CaP) and coated with a pH-responsive polymer to provide the enteric formulation of iron-ceria nanotablets (CFNT). CFNT exhibits a marked alleviative efficacy in the dextran sodium sulfate (DSS)-induced enterocolitis model in vivo by modulating the pro-inflammatory behavior of innate immune cells including macrophages and neutrophils, promoting anti-inflammatory cytokine profiles, and downregulating key transcription factors of inflammatory pathways.
Collapse
Affiliation(s)
- Seungmin Baik
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Hyunmin Kim
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
- Department of Medical Science, BK21 Plus KUMS Graduate Program, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Yunjung Lee
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Taegyu Kang
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Kwangsoo Shin
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Changyeong Song
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Ok Kyu Park
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Byeonggeun Kang
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nohyun Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Advanced Materials Engineering, Kookmin University, Seoul, 02707, Republic of Korea
| | - Dokyoon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan, 15558, Republic of Korea
| | - Seung Hong Choi
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Center for Advanced Pharmaceutical Technology, HyeonTechNBio Inc., Seoul, 08826, Republic of Korea
| | - Seung Han Kim
- Department of Gastroenterology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Min Soh
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Advanced Pharmaceutical Technology, HyeonTechNBio Inc., Seoul, 08826, Republic of Korea
| | - Taeghwan Hyeon
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Chi Kyung Kim
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
- Department of Medical Science, BK21 Plus KUMS Graduate Program, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| |
Collapse
|
12
|
Tian Y, Ye Z, Wang X, Guan H, Liu W, Duan X, Liu Y, Zeng G, Liu H. MOF-818 Nanozyme Suppresses Calcium Oxalate Kidney Stones by Alleviating Oxidative Stress and Inflammatory Injury. Adv Healthc Mater 2024:e2401574. [PMID: 39171497 DOI: 10.1002/adhm.202401574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/02/2024] [Indexed: 08/23/2024]
Abstract
There remains a lack of effective drugs to alleviate the kidney stones caused by oxidative stress and inflammatory damage. The MOF-818 nanozyme is utilized to lessen the generation of reactive oxygen species (ROS) effectively, restore the membrane potential of mitochondria, regulate the cell cycle, decrease cell death, hinder the recruitment of macrophages, and mitigate the release of inflammatory factors in macrophages. These effects are attributed to the nanozyme's ability to mimic the enzyme properties of catalase (CAT) and superoxide dismutase (SOD). It is demonstrated that this nanozyme can reduce kidney calcium oxalate crystal deposition by reducing the renal injury caused by high concentration oxalate, upregulate the expression levels of SOD and CAT in tissues, downregulate adhesion proteins and inflammatory factor IL-6 and TNF-α, and promote the polarization of macrophages from M1 to M2 phenotype in the rat model induced by ethylene glycol. Overall, MOF-818 has the potential to effectively suppress oxidative stress and inflammatory harm caused by high levels of oxalate, hence lowering the likelihood of stone formation. MOF-818 nanozyme is also expected to be used as an alternative drug for the treatment of calcium oxalate kidney stones and provide an experimental theoretical basis for the development of new nanomedicines.
Collapse
Affiliation(s)
- Yuan Tian
- Guangdong Provincial Key Laboratory of Urological Diseases, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China
| | - Ziyu Ye
- Guangdong Provincial Key Laboratory of Urological Diseases, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China
| | - Xunrui Wang
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences, Beijing, 100083, P. R. China
| | - Hantian Guan
- Guangdong Provincial Key Laboratory of Urological Diseases, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China
| | - Weifeng Liu
- Guangdong Provincial Key Laboratory of Urological Diseases, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China
| | - Xiaolu Duan
- Guangdong Provincial Key Laboratory of Urological Diseases, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China
| | - Yang Liu
- Guangdong Provincial Key Laboratory of Urological Diseases, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China
| | - Guohua Zeng
- Guangdong Provincial Key Laboratory of Urological Diseases, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China
| | - Hongxing Liu
- Guangdong Provincial Key Laboratory of Urological Diseases, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China
| |
Collapse
|
13
|
Luo X, Jiao Q, Pei S, Zhou S, Zheng Y, Shao W, Xu K, Zhong W. A Photoactivated Self-Assembled Nanoreactor for Inducing Cascade-Amplified Oxidative Stress toward Type I Photodynamic Therapy in Hypoxic Tumors. Adv Healthc Mater 2024:e2401787. [PMID: 39101321 DOI: 10.1002/adhm.202401787] [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: 05/15/2024] [Revised: 07/22/2024] [Indexed: 08/06/2024]
Abstract
Type I photodynamic therapy (PDT) generates reactive oxygen species (ROS) through oxygen-independent photoreactions, making it a promising method for treating hypoxic tumors. However, the superoxide anion (O2∙-) generated usually exhibits a low oxidation capacity, restricting the antitumor efficacy of PDT in clinical practice. Herein, a photoactivated self-assembled nanoreactor (1-NBS@CeO2) is designed through integration of type I PDT and cerium oxide (CeO2) nanozymes for inducing cascade-amplified oxidative stress in hypoxic tumors. The nanoreactor is constructed though co-assembly of an amphiphilic peptide (1-NBS) and CeO2, giving well-dispersed spherical nanoparticles with enhanced superoxide dismutase (SOD)-like and peroxidase (POD)-like activities. Following light irradiation, 1-NBS@CeO2 undergoes type I photoreactions to generated O2∙-, which is further catalyzed by the nanoreactors, ultimately forming hypertoxic hydroxyl radical (∙OH) through cascade-amplified reactions. The PDT treatment using 1-NBS@CeO2 results in elevation of intracellular ROS and depletion of GSH content in A375 cells, thereby inducing mitochondrial dysfunction and triggering apoptosis and ferroptosis of tumor cells. Importantly, intravenous administration of 1-NBS@CeO2 alongside light irradiation showcases enhances antitumor efficacy and satisfactory biocompatibility in vivo. Together, the self-assembled nanoreactor facilitates cascade-amplified photoreactions for achieving efficacious type I PDT, which holds great promise in developing therapeutic modules towards hypoxic tumors.
Collapse
Affiliation(s)
- Xuan Luo
- Department of Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Qishu Jiao
- Department of Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Shicheng Pei
- Department of Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Shuyao Zhou
- Department of Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Yaxin Zheng
- Department of Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Weiyang Shao
- Department of Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Keming Xu
- Department of Chemistry, China Pharmaceutical University, Nanjing, 210009, China
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing, 210009, China
| | - Wenying Zhong
- Department of Chemistry, China Pharmaceutical University, Nanjing, 210009, China
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing, 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
14
|
Dong L, Liu M, Fang M, Lu Q, Li X, Ma Y, Zhao T. Nucleation-Inhibited Emulsion Interfacial Assembled Polydopamine Microvesicles as Artificial Antigen-Presenting Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400714. [PMID: 38593314 DOI: 10.1002/smll.202400714] [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: 01/29/2024] [Revised: 03/31/2024] [Indexed: 04/11/2024]
Abstract
Albeit microemulsion systems have emerged as efficient platforms for fabricating tunable nano/microstructures, lack of understanding on the emulsion-interfacial assembly hindered the control of fabrication. Herein, a nucleation-inhibited microemulsion interfacial assembly method is proposed, which deviates from conventional interfacial nucleation approaches, for the synthesis of polydopamine microvesicles (PDA MVs). These PDA MVs exhibit an approximate diameter of 1 µm, showcasing a pliable structure reminiscent of cellular morphology. Through modifications of antibodies on the surface of PDA MVs, their capacity as artificial antigen presentation cells is evaluated. In comparison to solid nanoparticles, PDA MVs with cell-like structures show enhanced T-cell activation, resulting in a 1.5-fold increase in CD25 expression after 1 day and a threefold surge in PD-1 positivity after 7 days. In summary, the research elucidates the influence of nucleation and interfacial assembly in microemulsion polymerization systems, providing a direct synthesis method for MVs and substantiating their effectiveness as artificial antigen-presenting cells.
Collapse
Affiliation(s)
- Lingkai Dong
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Minchao Liu
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Meng Fang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P. R. China
| | - Qianqian Lu
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Xingjin Li
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Yanming Ma
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| | - Tiancong Zhao
- School of Chemistry and Materials, Department of Chemistry, Laboratory of Advanced Materials and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
15
|
Xin Q, Zhang S, Sun S, Song N, Zhe Y, Tian F, Zhang S, Guo M, Zhang XD, Zhang J, Wang H, Zhang R. Multienzyme Active Nanozyme for Efficient Sepsis Therapy through Modulating Immune and Inflammation Inhibition. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36047-36062. [PMID: 38978477 DOI: 10.1021/acsami.4c04994] [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: 07/10/2024]
Abstract
Sepsis, a life-threatening condition caused by a dysregulated immune response to infection, leads to systemic inflammation, immune dysfunction, and multiorgan damage. Various oxidoreductases play a very important role in balancing oxidative stress and modulating the immune response, but they are stored inconveniently, environmentally unstable, and expensive. Herein, we develop multifunctional artificial enzymes, CeO2 and Au/CeO2 nanozymes, exhibiting five distinct enzyme-like activities, namely, superoxide dismutase, catalase, glutathione peroxidase, peroxidase, and oxidase. These artificial enzymes have been used for the biocatalytic treatment of sepsis via inhibiting inflammation and modulating immune responses. These nanozymes significantly reduce reactive oxygen species and proinflammatory cytokines, achieving multiorgan protection. Notably, CeO2 and Au/CeO2 nanozymes with enzyme-mimicking activities can be particularly effective in restoring immunosuppression and maintaining homeostasis. The redox nanozyme offers a promising dual-protective strategy against sepsis-induced inflammation and organ dysfunction, paving the way for biocatalytic-based immunotherapies for sepsis and related inflammatory diseases.
Collapse
Affiliation(s)
- Qi Xin
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Tianjin Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
| | - Shaofang Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Si Sun
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Nan Song
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin 300384, China
| | - Yadong Zhe
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Fangzhen Tian
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Shu Zhang
- Department of Neurosurgery and Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Meili Guo
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin 300384, China
| | - Xiao-Dong Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Jianning Zhang
- Department of Neurosurgery and Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hao Wang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Ruiping Zhang
- The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Taiyuan 030032, China
| |
Collapse
|
16
|
Chen G, Cui L, Luo P, Fang J, Xie X, Jiang C. A Reactive Oxygen Species "Sweeper" Based on Hollow Mesopore Cerium Oxide Nanospheres for Targeted and Anti-Inflammatory Management of Osteoarthritis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:34705-34719. [PMID: 38935462 DOI: 10.1021/acsami.4c06231] [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: 06/29/2024]
Abstract
Osteoarthritis (OA) is a progressive joint disorder characterized by sustained oxidative stress, chronic inflammation, and the degradation of cartilage. Despite extensive research on nanocarrier treatment strategies, the therapeutic efficacy remains limited due to the lack of satisfactory vehicles that can simultaneously exhibit excellent ROS scavenging capabilities and high drug loading capacity for effective nonsurgical management of OA. In this work, we propose an innovative strategy utilizing hollow mesoporous cerium oxide nanospheres coated with membranes derived from apoptotic chondrocytes as a reactive oxygen species "sweeper" for targeted and anti-inflammatory therapy of OA. The developed DEX@HMCeNs@M demonstrates superior drug loading capacity, notable antioxidant properties, favorable biocompatibility, and controlled drug release. By leveraging the camouflage provided by apoptotic chondrocyte membranes, the engineered DEX@HMCeNs@M, which bear natural "eat me" signals, can effectively mimic chondrocyte apoptotic bodies within the joints, thereby enabling targeted delivery of the anti-inflammatory drug DEX and subsequent controlled release triggered by the acidic environment of OA. Both in vitro and in vivo experiments validate the enhanced therapeutic efficacy of our DEX@HMCeNs@M sweeper, which operates through a synergistic mechanism involving scavenging of ROS overproduction, inhibition of inflammation, restoration of mitochondrial damage, and reduction of chondrocyte apoptosis. These findings underscore the potential and efficiency of our developed DEX@HMCeNs@M strategy as an encouraging interventional approach for the progressive treatment of OA.
Collapse
Affiliation(s)
- Guofei Chen
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, 89 Taoyuan Road, Nanshan District, Shenzhen , Guangdong 518000, China
| | - Lei Cui
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, 89 Taoyuan Road, Nanshan District, Shenzhen , Guangdong 518000, China
| | - Peng Luo
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, 89 Taoyuan Road, Nanshan District, Shenzhen , Guangdong 518000, China
| | - Jiarui Fang
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, 89 Taoyuan Road, Nanshan District, Shenzhen , Guangdong 518000, China
| | - Xiaoxiao Xie
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, 89 Taoyuan Road, Nanshan District, Shenzhen , Guangdong 518000, China
| | - Changqing Jiang
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, 89 Taoyuan Road, Nanshan District, Shenzhen , Guangdong 518000, China
| |
Collapse
|
17
|
Nguyen TL, Phan NM, Kim J. Administration of ROS-Scavenging Cerium Oxide Nanoparticles Simply Mixed with Autoantigenic Peptides Induce Antigen-Specific Immune Tolerance against Autoimmune Encephalomyelitis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:33106-33120. [PMID: 38906850 DOI: 10.1021/acsami.4c05428] [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: 06/23/2024]
Abstract
The scavenging ability of cerium oxide nanoparticles (CeNPs) for reactive oxygen species has been intensively studied in the field of catalysis. However, the immunological impact of these particles has not yet been thoroughly investigated, despite intensive research indicating that modulation of the reactive oxygen species could potentially regulate cell fate and adaptive immune responses. In this study, we examined the intrinsic capability of CeNPs to induce tolerogenic dendritic cells via their reactive oxygen species-scavenging effect when the autoantigenic peptides were simply mixed with CeNPs. CeNPs effectively reduced the intracellular reactive oxygen species levels in dendritic cells in vitro, leading to the suppression of costimulatory molecules as well as NLRP3 inflammasome activation, even in the presence of pro-inflammatory stimuli. Subcutaneously administrated PEGylated CeNPs were predominantly taken up by antigen-presenting cells in lymph nodes and to suppress cell maturation in vivo. The administration of a mixture of PEGylated CeNPs and myelin oligodendrocyte glycoprotein peptides, a well-identified autoantigen associated with antimyelin autoimmunity, resulted in the generation of antigen-specific Foxp3+ regulatory T cells in mouse spleens. The induced peripheral regulatory T cells actively inhibited the infiltration of autoreactive T cells and antigen-presenting cells into the central nervous system, ultimately protecting animals from experimental autoimmune encephalomyelitis when tested using a mouse model mimicking human multiple sclerosis. Overall, our findings reveal the potential of CeNPs for generating antigen-specific immune tolerance to prevent multiple sclerosis, opening an avenue to restore immune tolerance against specific antigens by simply mixing the well-identified autoantigens with the immunosuppressive CeNPs.
Collapse
Affiliation(s)
- Thanh Loc Nguyen
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Ngoc Man Phan
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of MetaBioHealth, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
18
|
Lin Y, Tang Y, Yi O, Zhu J, Su Z, Li G, Zhou H, Liu L, Liu B, Cai X. Graphene oxide quantum dots-loaded sinomenine hydrochloride nanocomplexes for effective treatment of rheumatoid arthritis via inducing macrophage repolarization and arresting abnormal proliferation of fibroblast-like synoviocytes. J Nanobiotechnology 2024; 22:383. [PMID: 38951875 PMCID: PMC11218134 DOI: 10.1186/s12951-024-02645-8] [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: 04/17/2024] [Accepted: 06/16/2024] [Indexed: 07/03/2024] Open
Abstract
The characteristic features of the rheumatoid arthritis (RA) microenvironment are synovial inflammation and hyperplasia. Therefore, there is a growing interest in developing a suitable therapeutic strategy for RA that targets the synovial macrophages and fibroblast-like synoviocytes (FLSs). In this study, we used graphene oxide quantum dots (GOQDs) for loading anti-arthritic sinomenine hydrochloride (SIN). By combining with hyaluronic acid (HA)-inserted hybrid membrane (RFM), we successfully constructed a new nanodrug system named HA@RFM@GP@SIN NPs for target therapy of inflammatory articular lesions. Mechanistic studies showed that this nanomedicine system was effective against RA by facilitating the transition of M1 to M2 macrophages and inhibiting the abnormal proliferation of FLSs in vitro. In vivo therapeutic potential investigation demonstrated its effects on macrophage polarization and synovial hyperplasia, ultimately preventing cartilage destruction and bone erosion in the preclinical models of adjuvant-induced arthritis and collagen-induced arthritis in rats. Metabolomics indicated that the anti-arthritic effects of HA@RFM@GP@SIN NPs were mainly associated with the regulation of steroid hormone biosynthesis, ovarian steroidogenesis, tryptophan metabolism, and tyrosine metabolism. More notably, transcriptomic analyses revealed that HA@RFM@GP@SIN NPs suppressed the cell cycle pathway while inducing the cell apoptosis pathway. Furthermore, protein validation revealed that HA@RFM@GP@SIN NPs disrupted the excessive growth of RAFLS by interfering with the PI3K/Akt/SGK/FoxO signaling cascade, resulting in a decline in cyclin B1 expression and the arrest of the G2 phase. Additionally, considering the favorable biocompatibility and biosafety, these multifunctional nanoparticles offer a promising therapeutic approach for patients with RA.
Collapse
Affiliation(s)
- Ye Lin
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yuanyuan Tang
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Ouyang Yi
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Junping Zhu
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Zhaoli Su
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Gejing Li
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Hua Zhou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, University of Chinese Medicine, Guangzhou, 510006, China
| | - Liang Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, University of Chinese Medicine, Guangzhou, 510006, China.
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, Hunan, China.
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine, Department of Rheumatology of First Hospital, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
19
|
Zhuang C, Sun R, Zhang Y, Zou Q, Zhou J, Dong N, Zhao X, Fu W, Geng X, Wang J, Li Q, Zhao RC. Treatment of Rheumatoid Arthritis Based on the Inherent Bioactivity of Black Phosphorus Nanosheets. Aging Dis 2024:AD.2024.0319. [PMID: 38913037 DOI: 10.14336/ad.2024.0319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that affects the living quality of patients, especially the elderly population. RA-related morbidity and mortality increase significantly with age, while current clinical drugs for RA are far from satisfactory and may have serious side effects. Therefore, the development of new drugs with higher biosafety and efficacy is demanding. Black phosphorus nanosheets (BPNSs) have been widely studied because of their excellent biocompatibility. Here, we focus on the inherent bioactivity of BPNSs, report the potential of BPNSs as a therapeutic drug for RA and elucidate the underlying therapeutic mechanism. We find that BPNSs inhibit autophagy at an early stage via the AMPK-mTOR pathway, switch the energy metabolic pathway to oxidative phosphorylation, increase intracellular ATP levels, suppress apoptosis, reduce inflammation and oxidative stress, and down-regulate senescence-associated secretory phenotype (SASP)-related genes in rheumatoid arthritis synovial fibroblasts (RA-SFs). Further, BPNSs induce the apoptosis of macrophages and promote their transition from the M1 to the M2 phenotype by regulating related cytokines. Significantly, the administration of BPNSs can alleviate key pathological features of RA in mice, revealing great therapeutic potential. This study provides a novel option for treating RA, with BPNSs emerging as a promising therapeutic candidate.
Collapse
Affiliation(s)
- Cheng Zhuang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Ruiqi Sun
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Yuchen Zhang
- School of Medicine, Shanghai University, Shanghai, China
| | - Qing Zou
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Jianxin Zhou
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Naijun Dong
- School of Life Sciences, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Xuyu Zhao
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Wenjun Fu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xiaoke Geng
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qian Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
- Cell Energy Life Sciences Group Co. LTD, Qingdao, China, 266200
| | - Robert Chunhua Zhao
- School of Life Sciences, Shanghai University, Shanghai, China
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
| |
Collapse
|
20
|
Li C, Sun Y, Xu W, Chang F, Wang Y, Ding J. Mesenchymal Stem Cells-Involved Strategies for Rheumatoid Arthritis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305116. [PMID: 38477559 PMCID: PMC11200100 DOI: 10.1002/advs.202305116] [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: 07/26/2023] [Revised: 12/13/2023] [Indexed: 03/14/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of the joints and bone destruction. Because of systemic administration and poor targeting, traditional anti-rheumatic drugs have unsatisfactory treatment efficacy and strong side effects, including myelosuppression, liver or kidney function damage, and malignant tumors. Consequently, mesenchymal stem cells (MSCs)-involved therapy is proposed for RA therapy as a benefit of their immunosuppressive and tissue-repairing effects. This review summarizes the progress of MSCs-involved RA therapy through suppressing inflammation and promoting tissue regeneration and predicts their potential clinical application.
Collapse
Affiliation(s)
- Chaoyang Li
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Yifu Sun
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Fei Chang
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
| | - Yinan Wang
- Department of BiobankDivision of Clinical ResearchThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of EducationThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
21
|
Zhou H, Zhang Z, Mu Y, Yao H, Zhang Y, Wang DA. Harnessing Nanomedicine for Cartilage Repair: Design Considerations and Recent Advances in Biomaterials. ACS NANO 2024; 18:10667-10687. [PMID: 38592060 DOI: 10.1021/acsnano.4c00780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Cartilage injuries are escalating worldwide, particularly in aging society. Given its limited self-healing ability, the repair and regeneration of damaged articular cartilage remain formidable challenges. To address this issue, nanomaterials are leveraged to achieve desirable repair outcomes by enhancing mechanical properties, optimizing drug loading and bioavailability, enabling site-specific and targeted delivery, and orchestrating cell activities at the nanoscale. This review presents a comprehensive survey of recent research in nanomedicine for cartilage repair, with a primary focus on biomaterial design considerations and recent advances. The review commences with an introductory overview of the intricate cartilage microenvironment and further delves into key biomaterial design parameters crucial for treating cartilage damage, including microstructure, surface charge, and active targeting. The focal point of this review lies in recent advances in nano drug delivery systems and nanotechnology-enabled 3D matrices for cartilage repair. We discuss the compositions and properties of these nanomaterials and elucidate how these materials impact the regeneration of damaged cartilage. This review underscores the pivotal role of nanotechnology in improving the efficacy of biomaterials utilized for the treatment of cartilage damage.
Collapse
Affiliation(s)
- Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Yi Zhang
- School of Integrated Circuit Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
- Center for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, Hong Kong SAR 999077, China
| |
Collapse
|
22
|
Zhou G, Xu R, Groth T, Wang Y, Yuan X, Ye H, Dou X. The Combination of Bioactive Herbal Compounds with Biomaterials for Regenerative Medicine. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 38481114 DOI: 10.1089/ten.teb.2024.0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Regenerative medicine aims to restore the function of diseased or damaged tissues and organs by cell therapy, gene therapy, and tissue engineering, along with the adjunctive application of bioactive molecules. Traditional bioactive molecules, such as growth factors and cytokines, have shown great potential in the regulation of cellular and tissue behavior, but have the disadvantages of limited source, high cost, short half-life, and side effects. In recent years, herbal compounds extracted from natural plants/herbs have gained increasing attention. This is not only because herbal compounds are easily obtained, inexpensive, mostly safe, and reliable, but also owing to their excellent effects, including anti-inflammatory, antibacterial, antioxidative, proangiogenic behavior and ability to promote stem cell differentiation. Such effects also play important roles in the processes related to tissue regeneration. Furthermore, the moieties of the herbal compounds can form physical or chemical bonds with the scaffolds, which contributes to improved mechanical strength and stability of the scaffolds. Thus, the incorporation of herbal compounds as bioactive molecules in biomaterials is a promising direction for future regenerative medicine applications. Herein, an overview on the use of bioactive herbal compounds combined with different biomaterial scaffolds for regenerative medicine application is presented. We first introduce the classification, structures, and properties of different herbal bioactive components and then provide a comprehensive survey on the use of bioactive herbal compounds to engineer scaffolds for tissue repair/regeneration of skin, cartilage, bone, neural, and heart tissues. Finally, we highlight the challenges and prospects for the future development of herbal scaffolds toward clinical translation. Overall, it is believed that the combination of bioactive herbal compounds with biomaterials could be a promising perspective for the next generation of regenerative medicine.
Collapse
Affiliation(s)
- Guoying Zhou
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruojiao Xu
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Thomas Groth
- Department of Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Yanying Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xingyu Yuan
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hua Ye
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
- Oxford Suzhou Centre for Advanced Research, University of Oxford, Suzhou, China
| | - Xiaobing Dou
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
23
|
Ju Y, Yang P, Liu X, Qiao Z, Shen N, Lei L, Fang B. Microenvironment Remodeling Self-Healing Hydrogel for Promoting Flap Survival. Biomater Res 2024; 28:0001. [PMID: 38390027 PMCID: PMC10882600 DOI: 10.34133/bmr.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/13/2023] [Indexed: 02/24/2024] Open
Abstract
Random flap grafting is a routine procedure used in plastic and reconstructive surgery to repair and reconstruct large tissue defects. Flap necrosis is primarily caused by ischemia-reperfusion injury and inadequate blood supply to the distal flap. Ischemia-reperfusion injury leads to the production of excessive reactive oxygen species, creating a pathological microenvironment that impairs cellular function and angiogenesis. In this study, we developed a microenvironment remodeling self-healing hydrogel [laminarin-chitosan-based hydrogel-loaded extracellular vesicles and ceria nanozymes (LCH@EVs&CNZs)] to improve the flap microenvironment and synergistically promote flap regeneration and survival. The natural self-healing hydrogel (LCH) was created by the oxidation laminarin and carboxymethylated chitosan via a Schiff base reaction. We loaded this hydrogel with CNZs and EVs. CNZs are a class of nanomaterials with enzymatic activity known for their strong scavenging capacity for reactive oxygen species, thus alleviating oxidative stress. EVs are cell-secreted vesicular structures containing thousands of bioactive substances that can promote cell proliferation, migration, differentiation, and angiogenesis. The constructed LCH@EVs&CNZs demonstrated a robust capacity for scavenging excess reactive oxygen species, thereby conferring cellular protection in oxidative stress environments. Moreover, these constructs notably enhance cell migration and angiogenesis. Our results demonstrate that LCH@EVs&CNZs effectively remodel the pathological skin flap microenvironment and marked improve flap survival. This approach introduces a new therapeutic strategy combining microenvironmental remodeling with EV therapy, which holds promise for promoting flap survival.
Collapse
Affiliation(s)
- Yikun Ju
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Pu Yang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiangjun Liu
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zhihua Qiao
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Naisi Shen
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Lanjie Lei
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang 310015, China
| | - Bairong Fang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
24
|
Fu H, Guo Y, Fang W, Wang J, Hu P, Shi J. Anti-Acidification and Immune Regulation by Nano-Ceria-Loaded Mg-Al Layered Double Hydroxide for Rheumatoid Arthritis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307094. [PMID: 38064119 PMCID: PMC10853726 DOI: 10.1002/advs.202307094] [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: 09/26/2023] [Revised: 11/14/2023] [Indexed: 02/10/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease featuring an abnormal immune microenvironment and resultant accumulation of hydrogen ions (H+ ) produced by activated osteoclasts (OCs). Currently, clinic RA therapy can hardly achieve sustained or efficient therapeutic outcomes due to the failures in generating sufficient immune modulation and manipulating the accumulation of H+ that deteriorates bone damage. Herein, a highly effective immune modulatory nanocatalytic platform, nanoceria-loaded magnesium aluminum layered double hydroxide (LDH-CeO2 ), is proposed for enhanced immune modulation based on acid neutralization and metal ion inherent bioactivity. Specifically, the mild alkaline LDH initiates significant M2 repolarization of macrophages triggered by the elevated antioxidation effect of CeO2 via neutralizing excessive H+ in RA microenvironment, thus resulting in the efficient recruitment of regulatory T cell (Treg) and suppressions on T helper 17 cell (Th 17) and plasma cells. Moreover, the osteogenic activity is stimulated by the Mg ion released from LDH, thereby promoting the damaged bone healing. The encouraging therapeutic outcomes in adjuvant-induced RA model mice demonstrate the high feasibility of such a therapeutic concept, which provides a novel and efficient RA therapeutic modality by the immune modulatory and bone-repairing effects of inorganic nanocatalytic material.
Collapse
Affiliation(s)
- Hao Fu
- Shanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
| | - Yuedong Guo
- Platform of Nanomedicine TranslationShanghai Tenth People's HospitalMedical School of Tongji University38 Yun‐xin RoadShanghai200435P. R. China
| | - Wenming Fang
- Shanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
| | - Jiaxing Wang
- Department of OrthopaedicsShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai Jiao Tong UniversityShanghai200233P. R. China
| | - Ping Hu
- Shanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Platform of Nanomedicine TranslationShanghai Tenth People's HospitalMedical School of Tongji University38 Yun‐xin RoadShanghai200435P. R. China
| | - Jianlin Shi
- Shanghai Institute of CeramicsChinese Academy of SciencesResearch Unit of Nanocatalytic Medicine in Specific Therapy for Serious DiseaseChinese Academy of Medical Sciences (2021RU012)Shanghai200050P. R. China
- Platform of Nanomedicine TranslationShanghai Tenth People's HospitalMedical School of Tongji University38 Yun‐xin RoadShanghai200435P. R. China
| |
Collapse
|
25
|
Phillips R. Nanohybrid therapy hits multiple arthritis targets. Nat Rev Rheumatol 2024; 20:4. [PMID: 38062138 DOI: 10.1038/s41584-023-01064-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
|
26
|
Fang RH, Zhang L. Biohybrid nanoparticles for treating arthritis. NATURE NANOTECHNOLOGY 2023; 18:1387-1388. [PMID: 37884659 DOI: 10.1038/s41565-023-01503-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Affiliation(s)
- Ronnie H Fang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA, USA
| | - Liangfang Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|