1
|
Deng H, Qu Y, Chu B, Luo T, Pan M, Yuan L, Mo D, Bei Z, Yang T, Li X, Lu Y, Qian Z. Macrophage membrane-biomimetic ROS-responsive platinum nanozyme clusters for acute kidney injury treatment. Biomaterials 2025; 317:123072. [PMID: 39798243 DOI: 10.1016/j.biomaterials.2024.123072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/15/2025]
Abstract
Acute kidney injury (AKI) is a common clinical syndrome characterized by the rapid loss of renal filtration function. No standard therapeutic agent option is currently available. The development and progression of AKI is a continuous and dynamical pathological process. Oxidative stress and inflammatory responses are the primary influencing factors. Here, we developed a biomimetic nano-system (MM-PtNCs) with reactive oxygen species (ROS)-responsive platinum nanozyme clusters (PtNCs) wrapped in macrophage membrane (MM) to alleviate AKI by modulating oxidative stress and inflammation. The inflammatory cytokines receptors retained on MM surface allowed the biomimetic nano-system to target renal inflammation and neutralize these pro-inflammatory cytokines to ameliorate inflammation. PtNCs exhibit free radical scavenging-ability and catalase (CAT)-like activity to scavenge ROS and regulate the oxidative stress situations both in injured cells and tissues. Meanwhile, it could responsively dissociate into ultrasmall platinum nanoparticles under AKI-specific ROS conditions to eliminate ROS and eventually excreted through the renal filtration system. In a mouse model of ischemia/reperfusion-induced AKI, systemic injection of MM-PtNCs significantly reduced renal damage and restored kidney function. Additionally, MM-PtNCs effectively prevented the progression of AKI to chronic kidney disease. In conclusion, MM-PtNCs may propose a multi-faceted regulatory approach for clinical AKI treatment.
Collapse
Affiliation(s)
- Hanzhi Deng
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Ying Qu
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Bingyang Chu
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Tianying Luo
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Meng Pan
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Liping Yuan
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Dong Mo
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Zhongwu Bei
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Tingyu Yang
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xicheng Li
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Yi Lu
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Zhiyong Qian
- Department of Biotherapy and Department of Hematology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
2
|
Zhang N, Zhang X, Li J, Li B, Wang S, Liu X, Meng H, Zhu M. Development of Akkermansia Muciniphila Membrane-Coated Mesoporous Silica Nanoparticles with a Cerium Oxide Core for Inflammatory Bowel Disease Treatment. Adv Healthc Mater 2025; 14:e2405159. [PMID: 40181608 DOI: 10.1002/adhm.202405159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/17/2025] [Indexed: 04/05/2025]
Abstract
While convenient for patient compliance, the efficacy of oral treatments for inflammatory bowel disease (IBD) is often compromised by the dynamic and harsh chemical environment of the gastrointestinal tract, presenting challenges for effective therapeutic management. The pathological complexity of IBD frequently involves multiple factors such as oxidative stress, immune dysregulation, gut microbiome abnormality, and inadequate drug bioavailability, among others. To address these challenges, this project develops an oral nanomedicine platform based on mesoporous silica nanoparticles with a cerium oxide core, further coated with an outer membrane (OM) derived from Akkermansia muciniphila (Akk), a beneficial bacteria naturally present in the human gut. This novel nanocomplex, termed "OM-CeMeso," is evaluated for its enhanced stability, reactive oxygen species (ROS) scavenging capacity, and ability to restore microbiota homeostasis. This project demonstrates that the silica-based nanomaterials' acid-resistant yet base-degradable properties significantly improved stability in a murine IBD model. The incorporation of cerium oxide nanoparticles (CeO2 NPs) added further benefits by enhancing ROS scavenging. Notably, the Akk-derived OM coat also increases the diversity and abundance of beneficial gut microbiota. These complementary and integrated functions lead to significant symptom alleviation in murine IBD models while avoiding any unwanted toxicity.
Collapse
Affiliation(s)
- Nianhua Zhang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450001, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xu Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Jiulong Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Baoyi Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Shuai Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450001, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xiao Liu
- Department of Gastroenterology, Beijing Hospital, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, National Center of Gerontology, Beijing, 100730, China
| | - Huan Meng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Motao Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| |
Collapse
|
3
|
Li P, Yuan W, Hu K. Porous CeO 2 nanozyme with visible-light-enhanced catalase-mimicking activities by ligand-to-metal charge transfer. iScience 2025; 28:112149. [PMID: 40201121 PMCID: PMC11978324 DOI: 10.1016/j.isci.2025.112149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/07/2025] [Accepted: 02/27/2025] [Indexed: 04/10/2025] Open
Abstract
Nanozymes are promising synthetic alternatives to natural enzymes owing to their unique physical and chemical properties, but improving their thermocatalytic activity often involves complex procedures. This study introduces a light-induced approach to enhance the catalytic activity of facilely prepared porous cerium oxide ( P - CeO 2 ) nanozymes. Under visible light irradiation, the catalytic efficiency ( k c a t / K m ) of the P - CeO 2 more than doubles compared to its thermocatalytic efficiency. Spectroscopic analyses reveal that this enhancement stems from a ligand-to-metal charge transfer (LMCT) band, arising from peroxide species adsorbed on the P - CeO 2 surface. As a practical demonstration, P-CeO2 exhibits visible-light enhanced scavenging of reactive oxygen species (ROS) in vitro. Overall, this study provides new mechanistic insights of using LMCT-induced visible light catalysis for the improvement of catalytic activity of light-responsive nanozymes.
Collapse
Affiliation(s)
- Pengju Li
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Wei Yuan
- Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, Institute of Optoelectronics, Fudan University, Shanghai 200438, China
| | - Ke Hu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| |
Collapse
|
4
|
Wang Z, Zhang C. Nanomaterials for targeted therapy of kidney diseases: Strategies and advances. Mater Today Bio 2025; 31:101534. [PMID: 39990736 PMCID: PMC11846943 DOI: 10.1016/j.mtbio.2025.101534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
The treatment and management of kidney diseases pose a significant global burden. Due to the presence of blood circulation barriers and glomerular filtration barriers, drug therapy for kidney diseases faces challenges such as poor renal targeting, short half-life, and severe systemic side effects, severely hindering therapeutic progress. Therefore, the research and development of kidney-targeted therapeutic agents is of great clinical significance. In recent years, the application of nanotechnology in the field of nephrology has shown potential for revolutionizing the diagnosis and treatment of kidney diseases. Carefully designed nanomaterials can exhibit optimal biological characteristics, influencing various aspects such as circulation, retention, targeting, and excretion. Rationally designing and modifying nanomaterials based on the anatomical structure and pathophysiological environment of the kidney to achieve highly specific kidney-targeted nanomaterials or nanodrug delivery systems is both feasible and promising. Based on the targeted therapy of kidney diseases, this review discusses the advantages and limitations of current nanomedicine in the targeted therapy of kidney diseases, and summarizes the application and challenges of current renal active/passive targeting strategies, in order to further promote the development of kidney-targeted nanomedicine through a preliminary summary of previous studies and future prospects.
Collapse
Affiliation(s)
- Zhiwen Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
5
|
Shi G, Lan S, Zhang Q, Wang J, Shu F, Hao Z, Chen T, Zhu M, Chen R, Chen J, Wu Z, Wu B, Zou Z, Li J. Molybdenum nanodots act as antioxidants for photothermal therapy osteoarthritis. Biomaterials 2025; 315:122909. [PMID: 39471714 DOI: 10.1016/j.biomaterials.2024.122909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/21/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
Osteoarthritis (OA) manifests as the degradation of cartilage and remodeling of subchondral bone. Restoring homeostasis within the joint is imperative for alleviating OA symptoms. Current interventions primarily target singular aspects, such as anti-aging, inflammation inhibition, free radical scavenging, and regeneration of cartilage and subchondral bone. Herein, we developed molybdenum nanodots (MNDs) as bionic photothermal nanomaterials to mimic the antioxidant synthase to concurrently protected cartilage and facilitate subchondral bone regeneration. With near-infrared (NIR) irradiation, MNDs effectively eliminate reactive oxygen and nitrogen species (ROS/RNS) from OA chondrocytes, thereby reversed mitochondrial dysfunction, mitigating chondrocyte senescence, and simultaneously suppresses inflammation, hence preserving the inherent homeostasis between cartilage matrix synthesis and degradation while circumventing safety concerns. RNA sequencing of OA chondrocytes treated with MNDs-NIR revealed the reinstatement of chondrocyte functionality, activation of antioxidant enzymes, anti-aging properties, and regulation of inflammation. NIR irradiation induces thermogenesis and synergistically promotes subchondral bone regeneration via MNDs, as validated through histological assessments and microcomputed tomography (Micro-CT) scans. MNDs-NIR effectively attenuate cellular senescence and inhibit inflammation in vivo, while also remodeling mitochondrial dynamics by upregulating fusion proteins and inhibiting fission proteins, thereby regulating the oxidative stress microenvironment. Additionally, MNDs-NIR exhibited remarkable therapeutic effects in alleviating articular cartilage degeneration in an OA mouse model, evidenced by a 1.67-fold reduction in subchondral bone plate thickness, an 88.57 % decrease in OARSI score, a 5.52-fold reduction in MMP13 expression, and a 6.80-fold increase in Col II expression. This novel disease-modifying approach for OA utilizing MNDs-NIR offers insight and a paradigm for improving mitochondrial dysfunction by regulating the accumulation of mitochondrial ROS and ultimately alleviating cellular senescence. Moreover, the dual-pronged therapeutic approach of MNDs-NIR, which addresses both cartilage erosion and subchondral bone lesions in OA, represents a highly promising strategy for managing OA.
Collapse
Affiliation(s)
- Guang Shi
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, China
| | - Shenghui Lan
- Department of Orthopaedics, The Eighth People's Hospital, Jiangsu University, Shanghai, 200235, China
| | - Qi Zhang
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, China
| | - Junwu Wang
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, China
| | - Feihong Shu
- Department of Endoscopy and Digestive System, Guizhou Provincial People's Hospital, Guiyang, 550499, China
| | - Zhuowen Hao
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, China
| | - Tianhong Chen
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, China
| | - Mengyue Zhu
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, China
| | - Renxin Chen
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, China
| | - Jiayao Chen
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, China
| | - Zijian Wu
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, China
| | - Bo Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Zhenwei Zou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Jingfeng Li
- Department of Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, China.
| |
Collapse
|
6
|
Roointan A, Xu R, Corrie S, Hagemeyer CE, Alt K. Nanotherapeutics in Kidney Disease: Innovations, Challenges, and Future Directions. J Am Soc Nephrol 2025; 36:500-518. [PMID: 39705082 PMCID: PMC11888965 DOI: 10.1681/asn.0000000608] [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: 08/28/2024] [Accepted: 12/17/2024] [Indexed: 12/22/2024] Open
Abstract
The treatment and management of kidney diseases present a significant global challenge, affecting over 800 million individuals and necessitating innovative therapeutic strategies that transcend symptomatic relief. The application of nanotechnology to therapies for kidney diseases, while still in its early stages, holds transformative potential for improving treatment outcomes. Recent advancements in nanoparticle-based drug delivery leverage the unique physicochemical properties of nanoparticles for targeted and controlled therapeutic delivery to the kidneys. Current research is focused on understanding the functional and phenotypic changes in kidney cells during both acute and chronic conditions, allowing for the identification of optimal target cells. In addition, the development of tailored nanomedicines enhances their retention and binding to key renal membranes and cell populations, ultimately improving localization, tolerability, and efficacy. However, significant barriers remain, including inconsistent nanoparticle synthesis and the complexity of kidney-specific targeting. To overcome these challenges, the field requires advanced synthesis techniques, refined targeting strategies, and the establishment of animal models that accurately reflect human kidney diseases. These efforts are critical for the clinical application of nanotherapeutics, which promise novel solutions for kidney disease management. This review evaluates a substantial body of in vivo research, highlighting the prospects, challenges, and opportunities presented by nanotechnology-mediated therapies and their potential to transform kidney disease treatment.
Collapse
Affiliation(s)
- Amir Roointan
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- NanoTheranostics Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Rong Xu
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Simon Corrie
- Department of Chemical and Biological Engineering, Monash University, Melbourne, Victoria, Australia
| | - Christoph E. Hagemeyer
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Karen Alt
- NanoTheranostics Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Wang Y, Zhong C, Ma X, Liu X, Zhang B, Min C, Zhang K, Wang X, Hou S, Zhao J, Wang J, Fang Y, Liu H, Ding H, Guo J, Cheng Y, Lu W. Ceria Nanoparticles Improve the Quality of Aging Oocytes in Mice by Enhancing ROS Scavenging Ability. ACS APPLIED BIO MATERIALS 2025; 8:992-1001. [PMID: 39957429 DOI: 10.1021/acsabm.4c01086] [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: 02/18/2025]
Abstract
If fertilization does not occur in a timely manner after ovulation, then that can lead to aging and a decline in relation to oocyte quality, which have detrimental effects on subsequent embryo development and offspring. Reactive oxygen species (ROS) induced oxidative stress is the main factor in the deterioration of oocyte aging. Herein, ceria nanoparticles (NPs) have been developed as ROS scavengers to improve the quality of aging oocytes. We found that ceria NPs could effectively inhibit senescence-induced morphological changes and lower ROS accumulation in the oocytes. In addition, ceria NPs reduced the abnormal spindle distribution and improved mitochondrial function. Ceria NPs also reduced oxidative stress in aging oocytes by increasing ROS scavenging capacity rather than inhibiting ROS generation capacity. Furthermore, ceria NPs increased the rate of blastocyst formation in aging oocytes. We conclude that ceria NPs can enhance the quality and development potential of aging oocytes by reducing the level of oxidative stress.
Collapse
Affiliation(s)
- Yuchan Wang
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Chengkun Zhong
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Xin Ma
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Xin Liu
- Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, College of Life Science, Jilin Agricultural University, Changchun 130118, China
| | - Biao Zhang
- Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, College of Life Science, Jilin Agricultural University, Changchun 130118, China
| | - Changguo Min
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Kaiyan Zhang
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Xinhai Wang
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Shengkui Hou
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Jing Zhao
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Jun Wang
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Yi Fang
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Hongyu Liu
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - He Ding
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Jing Guo
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| | - Yan Cheng
- Engineering Research Center of Bioreactor and Pharmaceutical Development, Ministry of Education, College of Life Science, Jilin Agricultural University, Changchun 130118, China
| | - Wenfa Lu
- Key Lab of the Animal Production, Product Quality, and Security, Ministry of Education, College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin 130118, China
- Jilin Provincial International Joint Research Center of Animal Breeding & Reproduction Technology,Changchun130117,China
| |
Collapse
|
8
|
Li C, Wang W, Shao J, Zhou S, Ji X, Xi Y, Xu Q, Huang Y, Wang J, Wan Y, Li Z. Biomimetic polydopamine loaded with janus kinase inhibitor for synergistic vitiligo therapy via hydrogel microneedles. J Nanobiotechnology 2025; 23:63. [PMID: 39885576 PMCID: PMC11780829 DOI: 10.1186/s12951-025-03119-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Both oxidative stress and autoimmune responses play crucial roles in the development of vitiligo. Under oxidative stress, the apoptotic melanocytes expose self-antigens and release high mobility group box 1 (HMGB1), triggering autoimmune activation and recruiting CD8+ T cells. This process further leads to the destruction of melanocytes, resulting in the lack of melanin granules. Additionally, the accumulated CD8+ T cells release interferon-γ (IFN-γ) to activate janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway in keratinocytes. Both oxidative stress and IFN-γ-JAK-STAT activation induce keratinocytes to express and release T cell chemotactic factors, exacerbating the process of vitiligo. Reducing the accumulation of CD8+ T cells by safeguarding melanocytes and keratinocytes from oxidative stress may be contemplated as a promising approach for vitiligo therapy. RESULTS In this study, we introduce a novel therapeutic agent called PDA-JAKi, which is capable of both eliminating oxidative stress and inhibiting T cell activation. Specifically, we have incorporated the janus kinase inhibitor (JAKi) tofacitinib into antioxidant polydopamine (PDA) nanoparticles, resulting in the formation of uniform PDA-JAKi nanodrug. PDA-JAKi effectively mitigates oxidative stress-induced apoptosis in melanocytes, reducing the antigen presentation and release of HMGB1. In addition, PDA-JAKi simultaneously attenuates oxidative stress and blocks the IFN-γ-JAK-STAT pathway to reduce the expression of C-X-C motif chemokine ligand 9/10/16 (CXCL9/10/16) in keratinocytes. We precisely deliver this therapeutic agent to the dermis using microneedle (MN) patches, aiming to enhance therapeutic efficacy compared to traditional drug administration methods. After PDA-JAKi MN treatment, the symptoms of vitiligo in mice are alleviated, and the affected areas regain pigmentation. Enhancements have been observed in the dermal thickness, the numbers of melanocytes and the content of melanin within the treated skin area. Moreover, there is a notable reduction in reactive oxygen species (ROS) level. Concurrently, substantial decreases were noted in CD8+ T cell infiltration, as well as the levels of IFN-γ and chemotactic factors CXCL9/10/16. CONCLUSIONS In summary, PDA-JAKi MN patches emerge as a promising therapeutic agent for vitiligo treatment.
Collapse
Affiliation(s)
- Chunying Li
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wenwen Wang
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Junyi Shao
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Sen Zhou
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiaolin Ji
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Youxia Xi
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Qiuyang Xu
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yuhan Huang
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jingle Wang
- Department of Medical Oncology, The Third Affiliated Hospital of Shanghai University, Wenzhou, 325000, China
| | - Yilin Wan
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| | - Zhiming Li
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
9
|
Zhao X, Qin Y, Li B, Wang Y, Liu J, Wang B, Zhao J, Yin J, Zhang L, Li J, Huang J, Chen K, Liu L, Wu Y. Genetically engineered biomimetic ATP-responsive nanozyme for the treatment of cardiac fibrosis. J Nanobiotechnology 2025; 23:10. [PMID: 39780203 PMCID: PMC11715444 DOI: 10.1186/s12951-024-03083-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cardiac fibrosis plays a critical role in the progression of various forms of heart disease, significantly increasing the risk of sudden cardiac death. However, currently, there are no therapeutic strategies available to prevent the onset of cardiac fibrosis. METHODS AND RESULTS Here, biomimetic ATP-responsive nanozymes based on genetically engineered cell membranes are adapted to specifically recognize activated cardiac fibroblasts (CFs) for the treatment of cardiac fibrosis. By fusing the anti-FAP CAR genetically engineered cell membrane to zeolitic imidazole frameworks-90 (zif-90) cores loaded with antioxidant nanozymes CeO2 and siCTGF (siRNA targeting CTGF), these nanoparticles, called FM@zif-90/Ce/siR NPs, are demonstrated to effectively reduce the accumulation of myofibroblasts and the formation of fibrotic tissue, while restoring cardiac function. CONCLUSIONS These findings demonstrate that the combination of CeO2 and siCTGF has a beneficial curative effect on cardiac fibrosis, with significant translational potential.
Collapse
Affiliation(s)
- Xueli Zhao
- Department of Biochemistry and Molecular Biology, Shaanxi Provincial Key Laboratory of Clinical Genetics, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuze Qin
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bowen Li
- Department of Biochemistry and Molecular Biology, Shaanxi Provincial Key Laboratory of Clinical Genetics, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yue Wang
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiao Liu
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bo Wang
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jia Zhao
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiaqi Yin
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lanlan Zhang
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jing Li
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Junzhe Huang
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Kun Chen
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Liwen Liu
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Yuanming Wu
- Department of Laboratory, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Biochemistry and Molecular Biology, Shaanxi Provincial Key Laboratory of Clinical Genetics, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
10
|
Sun J, Shen H, Dong J, Zhang J, Yue T, Zhang R. Melanin-Deferoxamine Nanoparticles Targeting Ferroptosis Mitigate Acute Kidney Injury via RONS Scavenging and Iron Ion Chelation. ACS APPLIED MATERIALS & INTERFACES 2025; 17:282-296. [PMID: 39705095 DOI: 10.1021/acsami.4c14815] [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: 12/22/2024]
Abstract
Rhabdomyolysis (RM)-induced acute kidney injury (AKI) involves the release of large amounts of iron ions from excess myoglobin in the kidneys, which mediates the overproduction of reactive species with the onset of iron overload via the Fenton reaction, thus inducing ferroptosis and leading to renal dysfunction. Unfortunately, there are no effective treatments for AKI other than supportive care. Herein, we developed a multifunctional nanoplatform (MPD) by covalently bonding melanin nanoparticles (MP NPs) to deferoxamine. The nanoplatform has good dispersion and physiological stability, excellent chelating performance to iron ions, and broad-spectrum reactive species scavenging activity. Furthermore, cellular experiments showed that the NPs possessed high biocompatibility, antiapoptotic activity, antioxidant properties, and strong scavenging capacity of Fe2+ to mitigate iron overload, protecting the intracellular mitochondria from oxidative stress. Meanwhile, the intrinsic photoacoustic imaging capability of melanin allows the real-time monitoring of MPD NPs' target uptake and metabolic behavior in healthy and AKI mice. Most importantly, MPD NPs led to downregulation of the antioxidant pathway by targeting ferroptosis, thus effectively rescuing renal function in vivo, mitigating oxidative stress and inflammatory responses, and inhibiting renal tubular cell apoptosis. The nanoplatform offers a novel therapeutic strategy for RM-induced AKI.
Collapse
Affiliation(s)
- Jinghua Sun
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Hao Shen
- Shanxi Medical University, Taiyuan 030001, China
| | - Jie Dong
- Shanxi Medical University, Taiyuan 030001, China
| | - Jin Zhang
- Shanxi Medical University, Taiyuan 030001, China
| | - Tao Yue
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan 030012, China
| |
Collapse
|
11
|
Ren Y, Wu F, Huo L, Wang X, Zhang Y, Fan M, Tan M, Zhao J, Cheng J, Zhao Z, Bao J. Switchable ROS generator and scavenger to prevent the cisplatin induced acute kidney injury and improve efficacy via synergistic chemodynamic/immune therapy. Mater Today Bio 2024; 29:101328. [PMID: 39569165 PMCID: PMC11576404 DOI: 10.1016/j.mtbio.2024.101328] [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: 09/05/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
Acute kidney injury (AKI) induced by cisplatin (DDP), which is accompanied with the generation of reactive oxygen species (ROS), is a severe side effect during treatment and restricts the application of DDP. In this study, we develop ultrasmall Mn3O4 nanozyme (UMON) with tumor microenvironment (TME) responsive ROS scavenging and generating as adjuvant to alleviate DDP induced AKI with improved efficacy. In kidney, UMON with superoxide dismutase and catalase activity acts as ROS scavenger to eliminate ROS generated by DDP, successfully protecting the renal cells/tissue and alleviating AKI during DDP treatment. Alternatively, UMON rapidly responses to the high GSH level in TME and release Mn2+ in tumor. This unique feature endows it to generate hydroxyl radicals (∙OH) through a Fenton-like reaction and deplete GSH in tumor cell and tissue, achieving high efficient chemodynamic therapy (CDT). More importantly, the Mn2+ successfully activates the cGAS-STING pathway, initiating the immune response and effectively inhibiting the tumor metastases. The synergistic CDT and immune therapy effectively improve the anti-tumor efficacy of DDP in vitro and in vivo. This study demonstrates that TME responsive ROS scavenger/generator shows the potential to reduce side effects of DDP while improve its therapeutic efficacy, providing a new avenue to achieve efficient chemotherapy and promoting the progress of clinical chemotherapy.
Collapse
Affiliation(s)
- Yanan Ren
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450001, China
| | - Fei Wu
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450001, China
| | - Linlin Huo
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Xiao Wang
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450001, China
| | - Yong Zhang
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450001, China
| | - Mengke Fan
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Mingya Tan
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Jiayi Zhao
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Jingliang Cheng
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhenghuan Zhao
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Jianfeng Bao
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
12
|
Liao R, Du X, Zhai Z, Wang Y, Li J, Long Y, Jiang Y, Zheng H. Screen-printed, biocompatible and ultrasensitive sensor for real-time reactive oxygen species detection in human sweat. Talanta 2024; 280:126774. [PMID: 39216419 DOI: 10.1016/j.talanta.2024.126774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/05/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Excessive or burst generation of reactive oxygen species (ROS) can induce oxidative stress, precipitating a range of critical illnesses, including cancers, Parkinson's disease and Ischemia-reperfusion injury. Conventional biological assays for ROS, involving discrete steps of capturing, labelling, and spectrometric detection, are complex and time-intensive. Moreover, their accuracy is substantially compromised by the short lifespan (microseconds to milliseconds) of ROS. Consequently, there is a pressing need for a rapid and efficient method that enables real-time detection. In this study, we have developed a printable, flexible ROS sensor based on a robust nanoenzyme composite by direct deposition of the paste onto a flexible polyethylene terephthalate (PET) substrate. This device demonstrated the fast and real-time responses to the hydrogen peroxide (mimetic agent) in the laboratory and to total ROS in sweat of an individual, exhibiting an outstanding current response to hydrogen peroxide across a broad concentration range of 0.01-10 mM, with a limit of detection (LOD) of 1.85 μM. The device's sensitivity to hydrogen peroxide (136.59 μA mM-1 cm-2), was found to be 1.5 to 10 times higher than that of sensors previously reported. Moreover, the IFRS device successfully identified instantaneous ROS levels in the sweat of adult males in vitro, with amperometric response increased 8 times after half an hour strenuous exercise, thereby exhibiting excellent selectivity, remarkable stability, and confirmed high biosafety. Overall, the IFRS provides a viable and practical solution for simple, expedited, and real-time ROS detection in the near future.
Collapse
Affiliation(s)
- Rui Liao
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Xiaosong Du
- State Key Laboratory of Electronic Thin Films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Zhiqiang Zhai
- State Key Laboratory of Electronic Thin Films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yang Wang
- State Key Laboratory of Electronic Thin Films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Jun Li
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53706, United States
| | - Yin Long
- State Key Laboratory of Electronic Thin Films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| | - Yadong Jiang
- State Key Laboratory of Electronic Thin Films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Heng Zheng
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, China.
| |
Collapse
|
13
|
He X, Peng L, Zhou L, Liu H, Hao Y, Li Y, Lv Z, Zeng B, Guo X, Guo R. A biphasic drug-releasing microneedle with ROS scavenging and angiogenesis for the treatment of diabetic ulcers. Acta Biomater 2024; 189:270-285. [PMID: 39362454 DOI: 10.1016/j.actbio.2024.09.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
Diabetic ulcers are one of the common complications in diabetic patients. Delayed wound healing is associated with persistent pro-inflammatory M1 polarization, reduced angiogenesis and increased reactive oxygen species (ROS) in the microenvironment. Wound healing consists of multiple phases and therefore requires treatment tailored to each phase. In this study, a biphasic drug-releasing microneedle (MN) was fabricated to achieve early ROS scavenging and late accelerated angiogenesis to promote wound healing. Vascular endothelial growth factor (VEGF) was first encapsulated in methacryloylated sulfonated chitosan (SCSMA) microspheres (V@MP), and then V@MP was loaded into hyaluronic acid (HA) microneedles along with cerium dioxide nanoparticles (CONPs). Rapid dissolution of HA rapidly releases the CONPs to clear ROS, whereas the V@MP stays in the wound. SCSMA slow degradation prolongs the release of VEGF, thereby promoting angiogenesis. In vitro and in vivo studies have shown that this biphasic drug-releasing smart microneedle improves cell proliferation and migration, effectively scavenges ROS, promotes angiogenesis and tissue regeneration, and synergistically promotes M2 macrophage polarization. It provides a new delivery mode for nano-enzymes and growth factors that could be multifunctional and synergistic in the treatment of diabetic ulcers. STATEMENT OF SIGNIFICANCE: In our study, we present a microneedle (V@MP/C@MN) that can release drugs biphasically, which showed good repair ability in diabetic ulcer model. Large amounts of CONPs were rapidly released to alleviate oxidative stress during the inflammation of the wound, and V@MP stayed in the wound for a long period of time to release VEGF and promote angiogenesis in the late stage of wound healing. The results indicated that V@MP/C@MN could promote cell proliferation and migration, effectively scavenge ROS, promote angiogenesis and tissue regeneration, and synergistically promote M2 macrophage polarization, which could play a multifunctional and synergistic role in the treatment of diabetic ulcers.
Collapse
Affiliation(s)
- Xinyue He
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Lianghong Peng
- Department of Ophthalmology, General Hospital of Southern Theater Command, PLA, Guangzhou 510010, China
| | - Liming Zhou
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Huiling Liu
- Head Department of Oral and Maxillofacial Surgery, Leiden University Medical Centre, Amsterdam, De Boelelaan 1117, the Netherlands
| | - Yifan Hao
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yuhan Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Zijin Lv
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Baohui Zeng
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China
| | - Xinmin Guo
- Department of Ultrasound, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, China.
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Key Laboratory of Regenerative Medicine of Ministry of Education, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
14
|
Qin W, Huang J, Zhang M, Xu M, He J, Liu Q. Nanotechnology-Based Drug Delivery Systems for Treating Acute Kidney Injury. ACS Biomater Sci Eng 2024; 10:6078-6096. [PMID: 39226188 PMCID: PMC11480945 DOI: 10.1021/acsbiomaterials.4c01385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Acute kidney injury (AKI) is a disease that is characterized by a rapid decline in renal function and has a relatively high incidence in hospitalized patients. Sepsis, renal hypoperfusion, and nephrotoxic drug exposure are the main causes of AKI. The major therapy measures currently include supportive treatment, symptomatic treatment, and kidney transplantation. These methods are supportive treatments, and their results are not satisfactory. Fortunately, many new treatments that markedly improve the AKI therapy efficiency are emerging. These include antioxidant therapy, ferroptosis therapy, anti-inflammatory therapy, autophagy therapy, and antiapoptotic therapy. In addition, the development of nanotechnology has further promoted therapeutic effects on AKI. In this review, we highlight recent advances in the development of nanocarriers for AKI drug delivery. Emphasis has been placed on the latest developments in nanocarrier modification and design. We also summarize the applications of different nanocarriers in AKI treatment. Finally, the advantages and challenges of nanocarrier applications in AKI are summarized, and several nanomedicines that have been approved for clinical trials to treat diverse kidney diseases are listed.
Collapse
Affiliation(s)
- Wanbing Qin
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Jiaqi Huang
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Manting Zhang
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Mingwei Xu
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Junbing He
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Qinghua Liu
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
- Department
of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 Guangdong, China
- NHC Key
Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong
Provincial Key Laboratory of Nephrology, Guangzhou, 510080 Guangdong, China
| |
Collapse
|
15
|
Yang Y, Du J, Gan J, Song X, Shu J, An C, Lu L, Wei H, Che J, Zhao X. Neutrophil-Mediated Nanozyme Delivery System for Acute Kidney Injury Therapy. Adv Healthc Mater 2024; 13:e2401198. [PMID: 38899383 DOI: 10.1002/adhm.202401198] [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: 03/31/2024] [Revised: 05/31/2024] [Indexed: 06/21/2024]
Abstract
Reactive oxygen species (ROS) scavenging of nanozymes toward acute kidney injury (AKI) is a current promising strategy, however, the glomerular filtration barrier (GFB) limits their application for treating kidney related diseases. Here, a neutrophil-mediated delivery system able to hijack neutrophil to transport nanozyme-loaded cRGD-liposomes to inflamed kidney for AKI treatment by cRGD targeting integrin αvβ1 is reported. The neutrophil-mediated nanozyme delivery system demonstrated great antioxidant and anti-apoptosis ability in HK-2 and NRK-52E cell lines. Moreover, in ischemia-reperfusion (I/R) induced AKI mice, a single dose of LM@cRGD-LPs 12 h post-ischemia significantly reduces renal function indicators, alleviates renal pathological changes, and inhibits apoptosis of renal tubular cells and the expression of renal tubular injured marker, thus remarkably reducing the damage of AKI. Mechanistically, the treatment of LM@cRGD-LPs markedly inhibits the process of Nrf2 to the nucleus and reduces the expression of the downstream HO-1, achieves a 99.51% increase in renal tissue Nrf2 levels, and an 86.31% decrease in HO-1 levels after LM@cRGD-LPs treatment. In short, the strategy of neutrophil-mediated nanozyme delivery system hold great promise as a potential therapy for AKI or other inflammatory diseases.
Collapse
Affiliation(s)
- Yu Yang
- Department of Andrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
| | - Jiang Du
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Jingjing Gan
- Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Xiang Song
- Department of Andrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
| | - Jiaxin Shu
- Department of Andrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
| | - Chaoli An
- Department of Andrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Li Lu
- Department of Andrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Hui Wei
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Junyi Che
- Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Xiaozhi Zhao
- Department of Andrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
- Department of Andrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| |
Collapse
|
16
|
Wu Y, Wu Q, Fan X, Yang L, Zou L, Liu Q, Shi G, Yang X, Tang K. Study on chitosan/gelatin hydrogels containing ceria nanoparticles for promoting the healing of diabetic wound. J Biomed Mater Res A 2024; 112:1532-1547. [PMID: 38501727 DOI: 10.1002/jbm.a.37701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
Chronic inflammation at diabetic wound sites results in the uncontrolled accumulation of pro-inflammatory factors and reactive oxygen species (ROS), which impedes cell proliferation and delays wound healing. To promote the healing of diabetic wounds, chitosan/gelatin hydrogels containing ceria nanoparticles (CNPs) of various sizes were created in the current study. CNPs' efficacy in removingO 2 • - , •OH, and H2O2 was demonstrated, and the scavenging ability of CNPs of varying sizes was compared. The in vitro experiments demonstrated that hydrogels containing CNPs could effectively protect cells from ROS-induced damage and facilitate mouse fibroblast migration. Furthermore, during the treatment of diabetic wounds in vivo, hydrogels containing CNPs exhibited anti-inflammatory activity and could reduce the expression of the pro-inflammatory factors TNF-α (above 30%), IL-6 (above 90%), and IL-1β (above 80%), and effectively promote wound closure (above 80%) by inducing re-epithelialization, collagen deposition, and angiogenesis. In addition, the biological properties and therapeutic effects of hydrogels containing CNPs of various sizes were compared and discussed. The finding revealed that hydrogels with 4 nm CNPs exhibited more significant biological properties and had implications for diabetic wound treatment.
Collapse
Affiliation(s)
- Yonghui Wu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, China
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, China
| | - Qianqian Wu
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, China
| | - Xialian Fan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lu Yang
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, China
| | - Ling Zou
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, China
| | - Qingshan Liu
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, China
| | - Guangyou Shi
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, China
| | - Xiaochao Yang
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, China
| | - Keyong Tang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Jin W, Xie X, Shen S, Zhou X, Wang S, Zhang L, Su X. Ultrasmall polyvinylpyrrolidone-modified iridium nanoparticles with antioxidant and anti-inflammatory activity for acute pancreatitis alleviation. J Biomed Mater Res A 2024; 112:988-1003. [PMID: 38318924 DOI: 10.1002/jbm.a.37679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/29/2023] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
Acute pancreatitis (AP) is a common and serious acute inflammatory disease with high severity rate and mortality. Inflammation and oxidative stress play an extremely important role in the development of AP disease. Polyvinylpyrrolidone-modified iridium nanoparticles (IrNP-PVP) have multienzyme mimetic activity, and the aim of this article is to discuss the therapeutic alleviative effects of the ultrasmall nanozymes IrNP-PVP on AP through their antioxidant and anti-inflammatory effects. IrNP-PVP were proved to inhibit inflammation and scavenge reactive oxygen species (ROS) at the cellular level. The synthetic IrNP-PVP exhibit remarkable antioxidant and anti-inflammatory activities in the prevention and treatment of AP mice by establishing murine AP model, which can reduce the oxidative stress and inflammatory response. The results of this article indicated that the ultrasmall nanozymes IrNP-PVP effectively alleviate AP via scavenging ROS as well as suppressing inflammation both in vivo and in vitro, which might provide enormous promise for the AP management.
Collapse
Affiliation(s)
- Wenzhang Jin
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xueting Xie
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Shuqi Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xingjian Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Shunfu Wang
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Lijiang Zhang
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xiang Su
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
18
|
Yin M, Lei D, Liu Y, Qin T, Gao H, Lv W, Liu Q, Qin L, Jin W, Chen Y, Liang H, Wang B, Gao M, Zhang J, Lu J. NIR triggered polydopamine coated cerium dioxide nanozyme for ameliorating acute lung injury via enhanced ROS scavenging. J Nanobiotechnology 2024; 22:321. [PMID: 38849841 PMCID: PMC11162040 DOI: 10.1186/s12951-024-02570-w] [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: 04/10/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024] Open
Abstract
Acute lung injury (ALI) is a life threatening disease in critically ill patients, and characterized by excessive reactive oxygen species (ROS) and inflammatory factors levels in the lung. Multiple evidences suggest that nanozyme with diversified catalytic capabilities plays a vital role in this fatal lung injury. At present, we developed a novel class of polydopamine (PDA) coated cerium dioxide (CeO2) nanozyme (Ce@P) that acts as the potent ROS scavenger for scavenging intracellular ROS and suppressing inflammatory responses against ALI. Herein, we aimed to identify that Ce@P combining with NIR irradiation could further strengthen its ROS scavenging capacity. Specifically, NIR triggered Ce@P exhibited the most potent antioxidant and anti-inflammatory behaviors in lipopolysaccharide (LPS) induced macrophages through decreasing the intracellular ROS levels, down-regulating the levels of TNF-α, IL-1β and IL-6, up-regulating the level of antioxidant cytokine (SOD-2), inducing M2 directional polarization (CD206 up-regulation), and increasing the expression level of HSP70. Besides, we performed intravenous (IV) injection of Ce@P in LPS induced ALI rat model, and found that it significantly accumulated in the lung tissue for 6 h after injection. It was also observed that Ce@P + NIR presented the superior behaviors of decreasing lung inflammation, alleviating diffuse alveolar damage, as well as promoting lung tissue repair. All in all, it has developed the strategy of using Ce@P combining with NIR irradiation for the synergistic enhanced treatment of ALI, which can serve as a promising therapeutic strategy for the clinical treatment of ROS derived diseases as well.
Collapse
Affiliation(s)
- Mingjing Yin
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Doudou Lei
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yalan Liu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Tao Qin
- Department of Intensive Care Unit, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
| | - Huyang Gao
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Wenquan Lv
- Department of Emergency, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi, 530022, China
| | - Qianyue Liu
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Lian Qin
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Weiqian Jin
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yin Chen
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Hao Liang
- College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Bailei Wang
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Ming Gao
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Jianfeng Zhang
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China.
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China.
| |
Collapse
|
19
|
Zhang Y, Liu W, Wei G, Liu Q, Shao G, Gu X, Cui X, Zhou Z, Wang Y, Zhao S, Muhammad F, Li S, Li T, Du Y, Wei H. Bioinspired Nanozymes as Nanodecoys for Urinary Tract Infection Treatment. ACS NANO 2024; 18:9019-9030. [PMID: 38483200 DOI: 10.1021/acsnano.3c12783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Urinary tract infections (UTIs), common bacterial infections in communities and medical facilities, are mainly mediated by FimH. The glycan sites of the uromodulin protein play a crucial role in protecting against UTIs by interacting with FimH. A bioinspired approach using glycan-FimH interactions may effectively reduce bacteria through an antiadhesive mechanism, thereby curbing bacterial resistance. However, typical antiadhesive therapy alone fails to address the excessive reactive oxygen species and inflammatory response during UTIs. To bridge this gap, antioxidant nanozymes with antiadhesive ability were developed as nanodecoys to counter bacteria and inflammation. Specifically, ultrasmall dextran-coated ceria (DEC) was engineered to address UTIs, with dextran blocking FimH adhesion and ceria exhibiting anti-inflammatory properties. DECs, metabolizable by the kidneys, reduced bacterial content in the urinary tract, mitigating inflammation and tissue damage. In murine models, DECs successfully treated acute UTIs, repeated infections, and catheter-related UTIs. This dual approach not only highlights the potential of nanozymes for UTIs but also suggests applicability to other FimH-induced infections in the lungs and bowels, marking a significant advancement in nanozyme-based clinical approaches.
Collapse
Affiliation(s)
- Yihong Zhang
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Wanling Liu
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Gen Wei
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Quanyi Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Guoqiang Shao
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Xiang Gu
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Xiaomiao Cui
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Zijun Zhou
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yuting Wang
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Sheng Zhao
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Faheem Muhammad
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Sirong Li
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Tong Li
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yan Du
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Hui Wei
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu 210023, China
- NMPA Key Laboratory for Biomedical Optics, Hangzhou, Zhejiang 310018, China
| |
Collapse
|
20
|
Ba X, Ye T, Shang H, Tong Y, Huang Q, He Y, Wu J, Deng W, Zhong Z, Yang X, Wang K, Xie Y, Zhang Y, Guo X, Tang K. Recent Advances in Nanomaterials for the Treatment of Acute Kidney Injury. ACS APPLIED MATERIALS & INTERFACES 2024; 16:12117-12148. [PMID: 38421602 DOI: 10.1021/acsami.3c19308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Acute kidney injury (AKI) is a serious clinical syndrome with high morbidity, elevated mortality, and poor prognosis, commonly considered a "sword of Damocles" for hospitalized patients, especially those in intensive care units. Oxidative stress, inflammation, and apoptosis, caused by the excessive production of reactive oxygen species (ROS), play a key role in AKI progression. Hence, the investigation of effective and safe antioxidants and inflammatory regulators to scavenge overexpressed ROS and regulate excessive inflammation has become a promising therapeutic option. However, the unique physiological structure and complex pathological alterations in the kidneys render traditional therapies ineffective, impeding the residence and efficacy of most antioxidant and anti-inflammatory small molecule drugs within the renal milieu. Recently, nanotherapeutic interventions have emerged as a promising and prospective strategy for AKI, overcoming traditional treatment dilemmas through alterations in size, shape, charge, and surface modifications. This Review succinctly summarizes the latest advancements in nanotherapeutic approaches for AKI, encompassing nanozymes, ROS scavenger nanomaterials, MSC-EVs, and nanomaterials loaded with antioxidants and inflammatory regulator. Following this, strategies aimed at enhancing biocompatibility and kidney targeting are introduced. Furthermore, a brief discussion on the current challenges and future prospects in this research field is presented, providing a comprehensive overview of the evolving landscape of nanotherapeutic interventions for AKI.
Collapse
Affiliation(s)
- Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Ye
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wen Deng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zichen Zhong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoqi Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kangyang Wang
- Department of Urology, Wenchang People's Hospital, Wenchang 571300, Hainan Province, China
| | - Yabin Xie
- Department of Urology, Wenchang People's Hospital, Wenchang 571300, Hainan Province, China
| | - Yanlong Zhang
- GuiZhou University Medical College, Guiyang 550025, Guizhou Province, China
| | - Xiaolin Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
21
|
Singh S. Antioxidant nanozymes as next-generation therapeutics to free radical-mediated inflammatory diseases: A comprehensive review. Int J Biol Macromol 2024; 260:129374. [PMID: 38242389 DOI: 10.1016/j.ijbiomac.2024.129374] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/30/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
Recent developments in exploring the biological enzyme mimicking properties in nanozymes have opened a separate avenue, which provides a suitable alternative to the natural antioxidants and enzymes. Due to high and tunable catalytic activity, low cost of synthesis, easy surface modification, and good biocompatibility, nanozymes have garnered significant research interest globally. Several inorganic nanomaterials have been investigated to exhibit catalytic activities of some of the key natural enzymes, including superoxide dismutase (SOD), catalase, glutathione peroxidase, peroxidase, and oxidase, etc. These nanozymes are used for diverse biomedical applications including therapeutics, imaging, and biosensing in various cells/tissues and animal models. In particular, inflammation-related diseases are closely associated with reactive oxygen and reactive nitrogen species, and therefore effective antioxidants could be excellent therapeutics due to their free radical scavenging ability. Although biological enzymes and other artificial antioxidants could perform well in scavenging the reactive oxygen and nitrogen species, however, suffer from several drawbacks such as the requirement of strict physiological conditions for enzymatic activity, limited stability in the environment beyond their optimum pH and temperature, and high cost of synthesis, purification, and storage make then unattractive for broad-spectrum applications. Therefore, this review systematically and comprehensively presents the free radical-mediated evolution of various inflammatory diseases (inflammatory bowel disease, mammary gland fibrosis, and inflammation, acute injury of the liver and kidney, mammary fibrosis, and cerebral ischemic stroke reperfusion) and their mitigation by various antioxidant nanozymes in the biological system. The mechanism of free radical scavenging by antioxidant nanozymes under in vitro and in vivo experimental models and catalytic efficiency comparison with corresponding natural enzymes has also been presented.
Collapse
Affiliation(s)
- Sanjay Singh
- National Institute of Animal Biotechnology (NIAB), Opposite Journalist Colony, Near Gowlidoddy, Extended Q-City Road, Gachibowli, Hyderabad 500032, Telangana, India.
| |
Collapse
|
22
|
Kim YG, Lee Y, Lee N, Soh M, Kim D, Hyeon T. Ceria-Based Therapeutic Antioxidants for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2210819. [PMID: 36793245 DOI: 10.1002/adma.202210819] [Citation(s) in RCA: 56] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/20/2023] [Indexed: 06/18/2023]
Abstract
The growing interest in nanomedicine over the last 20 years has carved out a research field called "nanocatalytic therapy," where catalytic reactions mediated by nanomaterials are employed to intervene in disease-critical biomolecular processes. Among many kinds of catalytic/enzyme-mimetic nanomaterials investigated thus far, ceria nanoparticles stand out from others owing to their unique scavenging properties against biologically noxious free radicals, including reactive oxygen species (ROS) and reactive nitrogen species (RNS), by exerting enzyme mimicry and nonenzymatic activities. Much effort has been made to utilize ceria nanoparticles as self-regenerating antioxidative and anti-inflammatory agents for various kinds of diseases, given the detrimental effects of ROS and RNS therein that need alleviation. In this context, this review is intended to provide an overview as to what makes ceria nanoparticles merit attention in disease therapy. The introductory part describes the characteristics of ceria nanoparticles as an oxygen-deficient metal oxide. The pathophysiological roles of ROS and RNS are then presented, as well as their scavenging mechanisms by ceria nanoparticles. Representative examples of recent ceria-nanoparticle-based therapeutics are summarized by categorization into organ and disease types, followed by the discussion on the remaining challenges and future research directions.
Collapse
Affiliation(s)
- Young Geon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yunjung Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nohyun Lee
- School of Advanced Materials Engineering, Kookmin University, Seoul, 02707, Republic of Korea
| | - Min Soh
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Center for Advanced Pharmaceutical Technology, HyeonTechNBio, Inc., Seoul, 08826, Republic of Korea
| | - Dokyoon Kim
- Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan, 15588, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
23
|
Yang Y, Zhang JY, Ma ZJ, Wang SC, He P, Tang XQ, Yang CF, Luo X, Yang X, Li L, Zhang MC, Li Y, Yu JH. Visualization of therapeutic intervention for acute liver injury using low-intensity pulsed ultrasound-responsive phase variant nanoparticles. Biomater Sci 2024; 12:1281-1293. [PMID: 38252410 DOI: 10.1039/d3bm01423a] [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: 01/23/2024]
Abstract
Acute liver injury (ALI) is a highly fatal condition characterized by sudden massive necrosis of liver cells, inflammation, and impaired coagulation function. Currently, the primary clinical approach for managing ALI involves symptom management based on the underlying causes. The association between excessive reactive oxygen species originating from macrophages and acute liver injury is noteworthy. Therefore, we designed a novel nanoscale phase variant contrast agent, denoted as PFP@CeO2@Lips, which effectively scavenges reactive oxygen species, and enables visualization through low intensity pulsed ultrasound activation. The efficacy of the nanoparticles in scavenging excess reactive oxygen species from RAW264.7 and protective AML12 cells has been demonstrated through in vitro and in vivo experiments. Additionally, these nanoparticles have shown a protective effect against LPS/D-GalN attack in C57BL/6J mice. Furthermore, when exposed to LIPUS irritation, the nanoparticles undergo liquid-gas phase transition and enable ultrasound imaging.
Collapse
Affiliation(s)
- You Yang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Ju-Ying Zhang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Zi-Jun Ma
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Shi-Chun Wang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Ping He
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Xiao-Qing Tang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Chao-Feng Yang
- Department of Radiology, Affifiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xia Luo
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Xing Yang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Ling Li
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Mao-Chun Zhang
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Yang Li
- Department of Ultrasound, Yuechi People's Hospital, Guangan, 638300, Sichuan, China
- Department of Radiology, Affifiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Jin-Hong Yu
- Department of Ultrasound, Affifiliated Hospital of North Sichuan Medical College, Innovation Centre for Science and Technology of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
- Department of Ultrasound, Yuechi People's Hospital, Guangan, 638300, Sichuan, China
| |
Collapse
|
24
|
Pan J, Wu T, Chen L, Chen X, Zhang C, Wang Y, Li H, Guo J, Jiang W. A bimetallic nanozyme coordinated with quercetin for efficient radical scavenging and treatment of acute kidney injury. NANOSCALE 2024; 16:2955-2965. [PMID: 38247885 DOI: 10.1039/d3nr05255a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Acute kidney injury (AKI), characterized by tissue inflammation and oxidative damage, is a common and potentially life-threatening complication in patients. Quercetin, a natural antioxidant, possesses diverse pharmacological properties. However, limited stability and bioavailability hinder its clinical utilization. Moreover, the application of nanotechnology in antioxidant strategies for AKI treatment faces significant knowledge gaps. These gaps stem from limited understanding of the therapeutic mechanisms and renal clearance pathways. To tackle these issues, this study aims to develop an anti-oxidation nanozyme through the coordination of quercetin (Que) with a ruthenium (Ru) doped platinum (Pt) nanozyme (RuPt nanozyme). Compared to using Que or the RuPt nanozyme alone, the combined use of Que and the nanozyme led to enhanced antioxidant activities, especially in ABTS and DPPH free radical scavenging activities. Moreover, the modified nanozyme showed remarkable efficacy in scavenging reactive oxygen species and inhibiting apoptosis in a H2O2-induced cellular model. Additionally, the in vivo study showed that the coordination-modified nanozyme effectively alleviated glycerol- and cisplatin-induced AKI by inhibiting oxidative stress. Furthermore, this nanozyme exhibited superior therapeutic efficacy when compared to free quercetin and the RuPt nanozyme. In conclusion, the findings of our study suggest that the quercetin modified RuPt nanozyme (QCN) exhibits remarkable biocompatibility and holds significant promise for the therapeutic management of AKI.
Collapse
Affiliation(s)
- Jiangpeng Pan
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, P.R. China.
- Academy of Medical Sciences. Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Tingting Wu
- Academy of Medical Sciences. Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Lu Chen
- Department of Cardiovascular Diseases the First Clinical Medical College. Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Xiaoxi Chen
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Henan, 451464, China.
| | - Chao Zhang
- Academy of Medical Sciences. Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Yanyan Wang
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, P.R. China.
- Academy of Medical Sciences. Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Hao Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Henan, 451464, China.
| | - Jiancheng Guo
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, P.R. China.
| | - Wei Jiang
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, P.R. China.
- Academy of Medical Sciences. Zhengzhou University, Zhengzhou, Henan, P.R. China
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Henan, 451464, China.
| |
Collapse
|
25
|
Fang H, Xu S, Wang Y, Yang H, Su D. Endogenous stimuli-responsive drug delivery nanoplatforms for kidney disease therapy. Colloids Surf B Biointerfaces 2023; 232:113598. [PMID: 37866237 DOI: 10.1016/j.colsurfb.2023.113598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
Kidney disease is one of the most life-threatening health problems, affecting millions of people in the world. Commonly used steroids and immunosuppressants often fall exceptionally short of outcomes with inescapable systemic toxicity. With the booming research in nanobiotechnology, stimuli-responsive nanoplatform has come an appealing therapeutic strategy for kidney disease. Endogenous stimuli-responsive materials have shown profuse promise owing to their enhanced spatiotemporal control and precise to the location of the lesion. This review focuses on recent advances stimuli-responsive drug delivery nano-architectonics for kidney disease. First, a brief introduction of pathogenesis of kidney disease and pathological microenvironment were provided. Then, various endogenous stimulus involved in drug delivery nanoplatforms including pH, ROS, enzymes, and glucose were categorized based on the pathological mechanisms of kidney disease. Next, we separately summarized literature examples of endogenous stimuli-responsive nanomaterials, and outlined the design strategies and response mechanisms. Finally, the paper was concluded by discussing remaining challenges and future perspectives of endogenous stimuli-responsive drug delivery nanoplatform for expediting the speed of development and clinical applications.
Collapse
Affiliation(s)
- Hufeng Fang
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China.
| | - Shan Xu
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China
| | - Yu Wang
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China
| | - Hao Yang
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China
| | - Dan Su
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China.
| |
Collapse
|
26
|
Li L, Shen Y, Tang Z, Yang Y, Fu Z, Ni D, Cai X. Engineered nanodrug targeting oxidative stress for treatment of acute kidney injury. EXPLORATION (BEIJING, CHINA) 2023; 3:20220148. [PMID: 38264689 PMCID: PMC10742205 DOI: 10.1002/exp.20220148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 04/23/2023] [Indexed: 01/25/2024]
Abstract
Acute kidney injury (AKI) is a clinical syndrome characterized by a rapid decline in renal function, and is associated with a high risk of death. Many pathological changes happen in the process of AKI, including crucial alterations to oxidative stress levels. Numerous efforts have thus been made to develop effective medicines to scavenge excess reactive oxygen species (ROS). However, researchers have encountered several significant challenges, including unspecific biodistribution, high biotoxicity, and in vivo instability. To address these problems, engineered nanoparticles have been developed to target oxidative stress and treat AKI. This review thoroughly discusses the methods that empower nanodrugs to specifically target the glomerular filtration barrier and presents the latest achievements in engineering novel ROS-scavenging nanodrugs in clustered sections. The analysis of each study's breakthroughs and imperfections visualizes the progress made in developing effective nanodrugs with specific biodistribution and oxidative stress-targeting capabilities. This review fills the blank of a comprehensive outline over current progress in applying nanotechnology to treat AKI, providing potential insights for further research.
Collapse
Affiliation(s)
- Liwen Li
- Department of Ultrasound in MedicineShanghai Jiao Tong University School of Medicine Affiliated Sixth People's HospitalShanghaiPeople's Republic of China
| | - Yining Shen
- Department of Ultrasound in MedicineShanghai Jiao Tong University School of Medicine Affiliated Sixth People's HospitalShanghaiPeople's Republic of China
| | - Zhongmin Tang
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin‐MadisonWisconsinUSA
| | - Yuwen Yang
- Department of Ultrasound in MedicineShanghai Jiao Tong University School of Medicine Affiliated Sixth People's HospitalShanghaiPeople's Republic of China
| | - Zi Fu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Dalong Ni
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Xiaojun Cai
- Department of Ultrasound in MedicineShanghai Jiao Tong University School of Medicine Affiliated Sixth People's HospitalShanghaiPeople's Republic of China
| |
Collapse
|
27
|
Jin Y, Zhang J, Xu Y, Yi K, Li F, Zhou H, Wang H, Chan HF, Lao YH, Lv S, Tao Y, Li M. Stem cell-derived hepatocyte therapy using versatile biomimetic nanozyme incorporated nanofiber-reinforced decellularized extracellular matrix hydrogels for the treatment of acute liver failure. Bioact Mater 2023; 28:112-131. [PMID: 37250866 PMCID: PMC10209199 DOI: 10.1016/j.bioactmat.2023.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 04/07/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Reactive oxygen species (ROS)-associated oxidative stress, inflammation storm, and massive hepatocyte necrosis are the typical manifestations of acute liver failure (ALF), therefore specific therapeutic interventions are essential for the devastating disease. Here, we developed a platform consisting of versatile biomimetic copper oxide nanozymes (Cu NZs)-loaded PLGA nanofibers (Cu NZs@PLGA nanofibers) and decellularized extracellular matrix (dECM) hydrogels for delivery of human adipose-derived mesenchymal stem/stromal cells-derived hepatocyte-like cells (hADMSCs-derived HLCs) (HLCs/Cu NZs@fiber/dECM). Cu NZs@PLGA nanofibers could conspicuously scavenge excessive ROS at the early stage of ALF, and reduce the massive accumulation of pro-inflammatory cytokines, herein efficiently preventing the deterioration of hepatocytes necrosis. Moreover, Cu NZs@PLGA nanofibers also exhibited a cytoprotection effect on the transplanted HLCs. Meanwhile, HLCs with hepatic-specific biofunctions and anti-inflammatory activity acted as a promising alternative cell source for ALF therapy. The dECM hydrogels further provided the desirable 3D environment and favorably improved the hepatic functions of HLCs. In addition, the pro-angiogenesis activity of Cu NZs@PLGA nanofibers also facilitated the integration of the whole implant with the host liver. Hence, HLCs/Cu NZs@fiber/dECM performed excellent synergistic therapeutic efficacy on ALF mice. This strategy using Cu NZs@PLGA nanofiber-reinforced dECM hydrogels for HLCs in situ delivery is a promising approach for ALF therapy and shows great potential for clinical translation.
Collapse
Affiliation(s)
- Yuanyuan Jin
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Jiabin Zhang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Fenfang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Huicong Zhou
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| |
Collapse
|
28
|
Sahu A, Min K, Jeon SH, Kwon K, Tae G. Self-assembled hemin-conjugated heparin with dual-enzymatic cascade reaction activities for acute kidney injury. Carbohydr Polym 2023; 316:121088. [PMID: 37321716 DOI: 10.1016/j.carbpol.2023.121088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/16/2023] [Accepted: 06/01/2023] [Indexed: 06/17/2023]
Abstract
Nanozymes have prominent catalytic activities with high stability as a substitute for unstable and expensive natural enzymes. However, most nanozymes are metal/inorganic nanomaterials, facing difficulty in clinical translation due to their unproven biosafety and limited biodegradability issues. Hemin, an organometallic porphyrin, was newly found to possess superoxide dismutase (SOD) mimetic activity along with previously known catalase (CAT) mimetic activity. However, hemin has poor bioavailability due to its low water solubility. Therefore, a highly biocompatible and biodegradable organic-based nanozyme system with SOD/CAT mimetic cascade reaction activity was developed by conjugating hemin to heparin (HepH) or chitosan (CS-H). Between them, Hep-H formed a smaller (<50 nm) and more stable self-assembled nanostructure and even possessed much higher and more stable SOD and CAT activities as well as the cascade reaction activity compared to CS-H and free hemin. Hep-H also showed a better cell protection effect against reactive oxygen species (ROS) compared to CS-H and hemin in vitro. Furthermore, Hep-H was selectively delivered to the injured kidney upon intravenous administration at the analysis time point (24 h) and exhibited excellent therapeutic effects on an acute kidney injury model by efficiently removing ROS, reducing inflammation, and minimizing structural and functional damage to the kidney.
Collapse
Affiliation(s)
- Abhishek Sahu
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Sae Hyun Jeon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Kiyoon Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
29
|
Wu Q, Yang L, Zou L, Yang W, Liu Q, Zhang A, Cao J, Shi G, He J, Yang X. Small Ceria Nanoclusters with High ROS Scavenging Activity and Favorable Pharmacokinetic Parameters for the Amelioration of Chronic Kidney Disease. Adv Healthc Mater 2023; 12:e2300632. [PMID: 37167626 DOI: 10.1002/adhm.202300632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/08/2023] [Indexed: 05/13/2023]
Abstract
The over production of reactive oxygen species (ROS) plays a critical role in the progression of chronic kidney disease (CKD). Organic ROS scavengers currently used for CKD treatment do not satisfy low dosage and high efficiency requirements. Ceria nanomaterials featured with renewable ROS scavenging activity are potential candidates for CKD treatment. Herein, a method for the synthesis of ceria nanoclusters (NCs) featured with small size of ≈1.2 nm is reported. The synthesized NCs are modified by three hydrophilic ligands with different molecular weights, including succinic acid (SA), polyethylene glycol diacid 600 (PEG600), and polyethylene glycol diacid 2000 (PEG2000). The surface modified NCs exhibit excellent ROS scavenging activity due to the high Ce3+ /Ce4+ ratio in their crystal structures. Compared with bigger-sized ceria nanoparticles (NPs) (≈45 nm), NCs demonstrate smoother blood concentration-time curve, lower organ accumulation, and faster metabolic rate superiorities. The administration of NCs to CKD mice, especially PEG600 and PEG2000 modified NCs, can effectively inhibit oxidative stress, inflammation, renal fibrosis, and apoptosis in their kidneys. Due to these benefits, the constructed NCs can ameliorate the progression of CKD. These findings suggest that NCs is a potential redox nanomedicine for future clinical treatment of CKD.
Collapse
Affiliation(s)
- Qianqian Wu
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, 400038, China
| | - Lu Yang
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, 400038, China
| | - Ling Zou
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, 400038, China
| | - Wang Yang
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, 400038, China
| | - Qingshan Liu
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, 400038, China
| | - Anwei Zhang
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, 400038, China
| | - Jiang Cao
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, 400038, China
| | - Guangyou Shi
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, 400038, China
| | - Jian He
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, 400038, China
| | - Xiaochao Yang
- School of Biomedical Engineering and Medical Imaging, Army Medical University, Chongqing, 400038, China
| |
Collapse
|
30
|
Yang Y, Nan Y, Chen Q, Xiao Z, Zhang Y, Zhang H, Huang Q, Ai K. Antioxidative 0-dimensional nanodrugs overcome obstacles in AKI antioxidant therapy. J Mater Chem B 2023; 11:8081-8095. [PMID: 37540219 DOI: 10.1039/d3tb00970j] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Acute kidney injury (AKI) is a commonly encountered syndrome associated with various aetiologies and pathophysiological processes leading to enormous health risks and economic losses. In the absence of specific drugs to treat AKI, hemodialysis remains the primary clinical treatment for AKI patients. The revelation of the pathology opens new horizons for antioxidant therapy in the treatment of AKI. However, small molecule antioxidant drugs and common nanozymes have failed to challenge AKI due to their unsatisfactory drug properties and renal physiological barriers. 0-Dimensional (0D) antioxidant nanodrugs stand out at this time thanks to their small size and high performance. Recently, a number of research studies have been carried out around 0D nanodrugs for alleviating AKI, and their multi-antioxidant enzyme mimetic activities, smooth glomerular filtration barrier permeability and excellent biocompatibility have been investigated. Here, we comprehensively summarize recent advances in 0D nanodrugs for AKI antioxidant therapy. We classify these representative studies into three categories according to the characteristics of 0D nanomaterials, namely ultra-small metal nanodots, inorganic non-metallic quantum dots and polymer nanodots. We focus on the antioxidant mechanisms and their distribution in vivo in each inspiring work, and the purpose and ingenuity of each design are rigorously captured and described. Finally, we provide our reflections and prospects for 0D antioxidant nanodrugs in AKI treatment. This mini review provides unique insights and valuable clues in the design of 0D nanodrugs and other kidney absorbable drugs.
Collapse
Affiliation(s)
- Yuqi Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yayun Nan
- Geriatric Medical Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yuntao Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Huanan Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| |
Collapse
|
31
|
Wu J, Shang H, Zhang A, He Y, Tong Y, Huang Q, Liu X, Chen Z, Tang K. Antioxidant nanozymes in kidney injury: mechanism and application. NANOSCALE 2023; 15:13148-13171. [PMID: 37547960 DOI: 10.1039/d3nr01954c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Excessive production of reactive oxygen species (ROS) in the kidneys is involved in the pathogenesis of kidney diseases, such as acute kidney injury (AKI) and diabetic kidney disease (DKD), and is the main reason for the progression of kidney injury. ROS can easily lead to lipid peroxidation and damage the tubular epithelial cell membrane, proteins and DNA, and other molecules, which can trigger cellular oxidative stress. Effective scavenging of ROS can delay or halt the progression of kidney injury by reducing inflammation and oxidative stress. With the development of nanotechnology and an improved understanding of nanomaterials, more researchers are applying nanomaterials with antioxidant activity to treat kidney injury. This article reviews the detailed mechanism between ROS and kidney injury, as well as the applications of nanozymes with antioxidant effects based on different materials for various kidney injuries. To better guide the applications of antioxidant nanozymes in kidney injury and other inflammatory diseases, at the end of this review we also summarize the aspects of nanozymes that need to be improved. An in-depth understanding of the role played by ROS in the occurrence and progression of kidney injury and the mechanism by which antioxidant nanozymes reduce oxidative stress is conducive to improving the therapeutic effect in kidney injury and inflammation-related diseases.
Collapse
Affiliation(s)
- Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - An Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| |
Collapse
|
32
|
Zhang B, Li Q, Xu Q, Li B, Dong H, Mou Y. Polydopamine Modified Ceria Nanorods Alleviate Inflammation in Colitis by Scavenging ROS and Regulating Macrophage M2 Polarization. Int J Nanomedicine 2023; 18:4601-4616. [PMID: 37600119 PMCID: PMC10437713 DOI: 10.2147/ijn.s416049] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/03/2023] [Indexed: 08/22/2023] Open
Abstract
Background Inflammatory bowel disease (IBD) is closely related to higher intracellular oxidative stress. Therefore, developing a novel method to scavenge the harmful reactive oxygen species (ROS) and alleviate colon inflammation to treat IBD is a promising strategy. Methods CeO2@PDA-PEG (CeO2@PP) were synthesized by modifying ceria (CeO2) nanorods with polydopamine (PDA) and polyethylene glycol (PEG). The ROS scavenging ability of CeO2@PP was detected by using flow cytometry and confocal laser scanning microscope (CLSM). The anti-inflammatory ability of CeO2@PP was determined in vitro by treating lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. The biocompatibility of CeO2@PP was evaluated in vivo and in vitro. Moreover, the therapeutic effects of CeO2@PP in vivo were estimated in a dextran sulfate sodium salt (DSS)-induced colitis mouse model. Results Physicochemical property results demonstrated that PDA and PEG modification endowed CeO2 nanorods with excellent dispersibility and colloidal stability. CeO2@PP maintained superior enzyme-like activity, including superoxide dismutase (SOD) and catalase (CAT), indicating antioxidant ability. Moreover, in vitro results showed that CeO2@PP with PDA promotes LPS-induced RAW 264.7 macrophages into M2-type polarization. In addition, in vitro and in vivo results showed that CeO2@PP have great biocompatibility and biosafety. Animal experiments have shown that CeO2@PP have excellent anti-inflammatory effects against DSS-induced colitis and effectively alleviated intestinal mucosal injury. Conclusion The nanoplatform CeO2@PP possessed excellent antioxidant and anti-inflammatory properties for scavenging ROS and modulating macrophage polarization, which is beneficial for efficient colitis therapy.
Collapse
Affiliation(s)
- Bingqing Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Qiang Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Qinglin Xu
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Baochao Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Heng Dong
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Yongbin Mou
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| |
Collapse
|
33
|
Feng C, Xiong Z, Sun X, Zhou H, Wang T, Wang Y, Bai HX, Lei P, Liao W. Beyond antioxidation: Harnessing the CeO 2 nanoparticles as a renoprotective contrast agent for in vivo spectral CT angiography. Biomaterials 2023; 299:122164. [PMID: 37229807 DOI: 10.1016/j.biomaterials.2023.122164] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/29/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023]
Abstract
It is a challenging task to develop a contrast agent that not only provides excellent image contrast but also protects impaired kidneys from oxidative-related stress during angiography. Clinically approved iodinated CT contrast media are associated with potential renal toxicity, making it necessary to develop a renoprotective contrast agent. Here, we develop a CeO2 nanoparticles (NPs)-mediated three-in-one renoprotective imaging strategy, namely, i) renal clearable CeO2 NPs serve as a one-stone-two-birds antioxidative contrast agent, ii) low contrast media dose, and iii) spectral CT, for in vivo CT angiography (CTA). Benefiting from the merits of advanced sensitivity of spectral CT and K-edge energy of Cerium (Ce, 40.4 keV), an improved image quality of in vivo CTA is successfully achieved with a 10 times reduction of contrast agent dosage. In parallel, the sizes of CeO2 NPs and broad catalytic activities are suitable to be filtered via glomerulus thus directly alleviating the oxidative stress and the accompanying inflammatory injury of the kidney tubules. In addition, the low dosage of CeO2 NPs reduces the hypoperfusion stress of renal tubules induced by concentrated contrast agents used in angiography. This three-in-one renoprotective imaging strategy helps prevent kidney injury from being worsened during the CTA examination.
Collapse
Affiliation(s)
- Cai Feng
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zongling Xiong
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xianting Sun
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Hao Zhou
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China; Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Tianming Wang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ying Wang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Harrison X Bai
- Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Peng Lei
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China; Molecular Imaging Research Center of Central South University, Changsha, 410008, China.
| |
Collapse
|
34
|
Thao NTM, Do HDK, Nam NN, Tran NKS, Dan TT, Trinh KTL. Antioxidant Nanozymes: Mechanisms, Activity Manipulation, and Applications. MICROMACHINES 2023; 14:1017. [PMID: 37241640 PMCID: PMC10220853 DOI: 10.3390/mi14051017] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
Antioxidant enzymes such as catalase, superoxide dismutase, and glutathione peroxidase play important roles in the inhibition of oxidative-damage-related pathological diseases. However, natural antioxidant enzymes face some limitations, including low stability, high cost, and less flexibility. Recently, antioxidant nanozymes have emerged as promising materials to replace natural antioxidant enzymes for their stability, cost savings, and flexible design. The present review firstly discusses the mechanisms of antioxidant nanozymes, focusing on catalase-, superoxide dismutase-, and glutathione peroxidase-like activities. Then, we summarize the main strategies for the manipulation of antioxidant nanozymes based on their size, morphology, composition, surface modification, and modification with a metal-organic framework. Furthermore, the applications of antioxidant nanozymes in medicine and healthcare are also discussed as potential biological applications. In brief, this review provides useful information for the further development of antioxidant nanozymes, offering opportunities to improve current limitations and expand the application of antioxidant nanozymes.
Collapse
Affiliation(s)
- Nguyen Thi My Thao
- School of Medicine and Pharmacy, Tra Vinh University, Tra Vinh City 87000, Vietnam
| | - Hoang Dang Khoa Do
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ward 13, District 04, Ho Chi Minh City 70000, Vietnam
| | - Nguyen Nhat Nam
- Biotechnology Center, School of Agriculture and Aquaculture, Tra Vinh University, Tra Vinh City 87000, Vietnam
| | - Nguyen Khoi Song Tran
- College of Korean Medicine, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Republic of Korea;
| | | | - Kieu The Loan Trinh
- Department of BioNano Technology, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| |
Collapse
|
35
|
Zhou C, Zhang L, Xu Z, Sun T, Gong M, Liu Y, Zhang D. Self-Propelled Ultrasmall AuNPs-Tannic Acid Hybrid Nanozyme with ROS-Scavenging and Anti-Inflammatory Activity for Drug-Induced Liver Injury Alleviation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206408. [PMID: 36759965 DOI: 10.1002/smll.202206408] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/05/2023] [Indexed: 05/11/2023]
Abstract
Developing nanomedicines with superior reactive oxygen species (ROS) scavenging capability has emerged as a promising strategy in treating ROS-related diseases, for example, drug-induced liver injury. However, designing nanoscavengers with the self-propelling ability to scavenge ROS actively remains challenging. Here, a self-propelled silica-supported ultrasmall gold nanoparticles-tannic acid hybrid nanozyme (SAuPTB) is designed that can effectively alleviate acetaminophen (APAP)-induced liver injury by scavenging excessive ROS and regulating inflammation. SAuPTB exhibits multienzyme activity and displays significantly enhanced diffusion under hydrogen peroxide (H2 O2 ). This in vitro research shows that SAuPTB can effectively eliminate ROS, increasing the viability of H2 O2 -stimulated cells and reducing the cytotoxicity of APAP/H2 O2 -treated AML12 cells. The in vivo studies show that SAuPTB can accumulate at inflammatory sites in mouse liver, resulting in the decrease of alanine aminotransferase, aspartate aminotransferase, and ROS, reduction in pro-inflammatory cytokines and chemokines, hence reduced hepatocyte necrosis, liver injury, and mortality. Furthermore, SAuPTB activates the nuclear erythroid 2-related factor 2 pathway to upregulate antioxidative genes and reduce oxidative stress. Finally, the liver shows decreased high mobility group box 1 and F4/80+ macrophages, suggesting an anti-inflammatory response. This work provides a novel design strategy of nanozymes for ROS-related disease treatment.
Collapse
Affiliation(s)
- Chunyu Zhou
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Liang Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Zhongsheng Xu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Tao Sun
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Mingfu Gong
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Yun Liu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Dong Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| |
Collapse
|
36
|
Nanodrugs alleviate acute kidney injury: Manipulate RONS at kidney. Bioact Mater 2023; 22:141-167. [PMID: 36203963 PMCID: PMC9526023 DOI: 10.1016/j.bioactmat.2022.09.021] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/12/2022] [Accepted: 09/19/2022] [Indexed: 02/06/2023] Open
Abstract
Currently, there are no clinical drugs available to treat acute kidney injury (AKI). Given the high prevalence and high mortality rate of AKI, the development of drugs to effectively treat AKI is a huge unmet medical need and a research hotspot. Although existing evidence fully demonstrates that reactive oxygen and nitrogen species (RONS) burst at the AKI site is a major contributor to AKI progression, the heterogeneity, complexity, and unique physiological structure of the kidney make most antioxidant and anti-inflammatory small molecule drugs ineffective because of the lack of kidney targeting and side effects. Recently, nanodrugs with intrinsic kidney targeting through the control of size, shape, and surface properties have opened exciting prospects for the treatment of AKI. Many antioxidant nanodrugs have emerged to address the limitations of current AKI treatments. In this review, we systematically summarized for the first time about the emerging nanodrugs that exploit the pathological and physiological features of the kidney to overcome the limitations of traditional small-molecule drugs to achieve high AKI efficacy. First, we analyzed the pathological structural characteristics of AKI and the main pathological mechanism of AKI: hypoxia, harmful substance accumulation-induced RONS burst at the renal site despite the multifactorial initiation and heterogeneity of AKI. Subsequently, we introduced the strategies used to improve renal targeting and reviewed advances of nanodrugs for AKI: nano-RONS-sacrificial agents, antioxidant nanozymes, and nanocarriers for antioxidants and anti-inflammatory drugs. These nanodrugs have demonstrated excellent therapeutic effects, such as greatly reducing oxidative stress damage, restoring renal function, and low side effects. Finally, we discussed the challenges and future directions for translating nanodrugs into clinical AKI treatment. AKI is a common clinical acute syndrome with high morbidity and mortality but without effective clinical drug available. Hypoxia and accumulation of toxic substances are key pathological features of various heterogeneous AKI. Excessive RONS is the core of the pathological mechanism of AKI. The development of nanodrugs is expected to achieve successful treatment in AKI.
Collapse
|
37
|
Zhang L, Meng W, Chen X, Wu L, Chen M, Zhou Z, Chen Y, Yuan L, Chen M, Chen J, Shui P. Multifunctional Nanoplatform for Mild Microwave-Enhanced Thermal, Antioxidative, and Chemotherapeutic Treatment of Rheumatoid Arthritis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10341-10355. [PMID: 36790223 DOI: 10.1021/acsami.2c19198] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Rheumatoid arthritis (RA) is usually associated with excessive proliferation of M1-type proinflammatory macrophages, resulting in severe hypoxia and excess reactive oxygen species (ROS) in the joint cavity. Inhibiting M1-type proinflammatory macrophages and/or repolarizing them into M2 phenotype anti-inflammatory cells by alleviating hypoxia and scavenging ROS could be a promising strategy for RA treatment. In this work, a microwave-sensitive metal-organic framework of UiO-66-NH2 is constructed for coating a nanoenzyme of cerium oxide (CeO2) and loading with the drug celastrol (Cel) to give UiO-66-NH2/CeO2/Cel, which is ultimately wrapped with hyaluronic acid (HA) to form a nanocomposite UiO-66-NH2/CeO2/Cel@HA (UCCH). With the microwave-susceptible properties of UiO-66-NH2, the thermal effect of microwaves can eliminate the excessive proliferation of inflammatory cells. In addition, superoxide-like and catalase-like activities originating from CeO2 in UCCH are boosted to scavenge ROS and accelerate the decomposition of H2O2 to produce O2 under microwave irradiation. The nonthermal effect of microwaves could synergistically promote the repolarization of M1-type macrophages into the M2 phenotype. Accompanied by the release of the anti-RA chemotherapeutic drug Cel, UCCH can efficiently ameliorate RA in vitro and in vivo through microwave-enhanced multisynergistic effects. This strategy could inspire the design of other multisynergistic platforms enhanced by microwaves to exploit new treatment modalities in RA therapies.
Collapse
Affiliation(s)
- Lianying Zhang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wei Meng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaotong Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Libo Wu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Mingwa Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhaoxi Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yongjian Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lixia Yuan
- School of Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ming Chen
- The People's Hospital of Gaozhou, Maoming 525200, China
| | - Jinxiang Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Pixian Shui
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
38
|
Gao Y, Liu S, Zeng X, Guo Z, Chen D, Li S, Tian Z, Qu Y. Reduction of Reactive Oxygen Species Accumulation Using Gadolinium-Doped Ceria for the Alleviation of Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10414-10425. [PMID: 36802486 DOI: 10.1021/acsami.2c20492] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Atherosclerosis is a common cardiovascular disease with increasing morbidity and mortality. The pathogenesis of atherosclerosis is strongly related to endothelial dysfunction, which is induced by severe oxidative stress damage derived from reactive oxygen species (ROS). Thus, ROS plays a critical role in the pathogenesis and progression of atherosclerosis. In this work, we demonstrated that the gadolinium doping of CeO2 (Gd/CeO2) nanozymes as effective ROS scavengers delivered high performance for antiatherosclerosis. It was found that the chemical doping of Gd promoted the surface proportion of Ce3+ in the nanozymes and thereby enhanced the overall ROS scavenging ability. In vitro and in vivo experiments unambiguously showed that the Gd/CeO2 nanozymes efficiently scavenged harmful ROS at the cellular and histological levels. Further, Gd/CeO2 nanozymes were demonstrated to significantly reduce vascular lesions by reducing lipid accumulation in macrophage and decreasing inflammatory factor levels, thereby inhibiting the exacerbation of atherosclerosis. Moreover, Gd/CeO2 can serve as T1-weighted magnetic resonance imaging contrast agents, which can generate sufficient contrast to distinguish the location of plaque during living imaging. Through those efforts, Gd/CeO2 may serve as a potential diagnostic and treatment nanomedicine for the ROS-induced atherosclerosis.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Shihong Liu
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xinchun Zeng
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhixiong Guo
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Da Chen
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Sanzhong Li
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, China
| | - Zhimin Tian
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yongquan Qu
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
39
|
Administration of a single dose of lithium ameliorates rhabdomyolysis-associated acute kidney injury in rats. PLoS One 2023; 18:e0281679. [PMID: 36795689 PMCID: PMC9934413 DOI: 10.1371/journal.pone.0281679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/27/2023] [Indexed: 02/17/2023] Open
Abstract
Rhabdomyolysis is characterized by muscle damage and leads to acute kidney injury (AKI). Clinical and experimental studies suggest that glycogen synthase kinase 3β (GSK3β) inhibition protects against AKI basically through its critical role in tubular epithelial cell apoptosis, inflammation and fibrosis. Treatment with a single dose of lithium, an inhibitor of GSK3β, accelerated recovery of renal function in cisplatin and ischemic/reperfusion-induced AKI models. We aimed to evaluate the efficacy of a single dose of lithium in the treatment of rhabdomyolysis-induced AKI. Male Wistar rats were allocated to four groups: Sham, received saline 0.9% intraperitoneally (IP); lithium (Li), received a single IP injection of lithium chloride (LiCl) 80 mg/kg body weight (BW); glycerol (Gly), received a single dose of glycerol 50% 5 mL/kg BW intramuscular (IM); glycerol plus lithium (Gly+Li), received a single dose of glycerol 50% IM plus LiCl IP injected 2 hours after glycerol administration. After 24 hours, we performed inulin clearance experiments and collected blood / kidney / muscle samples. Gly rats exhibited renal function impairment accompanied by kidney injury, inflammation and alterations in signaling pathways for apoptosis and redox state balance. Gly+Li rats showed a remarkable improvement in renal function as well as kidney injury score, diminished CPK levels and an overstated decrease of renal and muscle GSK3β protein expression. Furthermore, administration of lithium lowered the amount of macrophage infiltrate, reduced NFκB and caspase renal protein expression and increased the antioxidant component MnSOD. Lithium treatment attenuated renal dysfunction in rhabdomyolysis-associated AKI by improving inulin clearance and reducing CPK levels, inflammation, apoptosis and oxidative stress. These therapeutic effects were due to the inhibition of GSK3β and possibly associated with a decrease in muscle injury.
Collapse
|
40
|
Wang Y, Jiang H, Zhang L, Yao P, Wang S, Yang Q. Nanosystems for oxidative stress regulation in the anti-inflammatory therapy of acute kidney injury. Front Bioeng Biotechnol 2023; 11:1120148. [PMID: 36845189 PMCID: PMC9949729 DOI: 10.3389/fbioe.2023.1120148] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Acute kidney injury (AKI) is a clinical syndrome that results from a rapid decline in renal structure or renal functional impairment with the main pathological feature of sublethal and lethal damage to renal tubular cells. However, many potential therapeutic agents cannot achieve the desired therapeutic effect because of their poor pharmacokinetics and short retention time in the kidneys. With the recent emergence and progress of nanotechnology, nanodrugs with unique physicochemical properties could prolong circulation time, enhance efficient targeted delivery, and elevate the accumulation of therapeutics that can cross the glomerular filtration barrier and indicate comprehensive application prospects in the prevention and treatment of AKI. In this review, various types of nanosystems (such as liposomes, polymeric nanosystems, inorganic nanoparticles and cell-derived extracellular vesicles) are designed and applied to improve the pharmacokinetics of drug formation, which could further relieve the burden on the kidneys caused by the final cumulative dose of drugs in conventional treatments. Moreover, the passive or active targeting effect of nanosystems can also reduce the total therapeutic dose and off-target adverse effects on other organs. Nanodelivery systems for treating AKI that alleviate oxidative stress-induced renal cell damage and regulate the inflammatory kidney microenvironment are summarized.
Collapse
Affiliation(s)
- Yue Wang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China,Center of Scientific Research, Chengdu Medical College, Chengdu, Sichuan, China
| | - Hong Jiang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China,Center of Scientific Research, Chengdu Medical College, Chengdu, Sichuan, China
| | - Longyao Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China,Center of Scientific Research, Chengdu Medical College, Chengdu, Sichuan, China
| | - Peng Yao
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Shaoqing Wang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China,*Correspondence: Shaoqing Wang, ; Qian Yang,
| | - Qian Yang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China,Center of Scientific Research, Chengdu Medical College, Chengdu, Sichuan, China,*Correspondence: Shaoqing Wang, ; Qian Yang,
| |
Collapse
|
41
|
Liu Y, Yan X, Wei H. Medical Nanozymes for Therapeutics. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
42
|
Si P, Lei J, Yang C, Zhang P, Li X, Zheng S, Li Q, Zhang J. Mesoporous Hollow Manganese Doped Ceria Nanoparticle for Effectively Prevention of Hepatic Ischemia Reperfusion Injury. Int J Nanomedicine 2023; 18:2225-2238. [PMID: 37131546 PMCID: PMC10149098 DOI: 10.2147/ijn.s400467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/04/2023] [Indexed: 05/04/2023] Open
Abstract
Introduction Hepatic ischemia-reperfusion injury (HIRI) is the main reason for liver dysfunction or failure after liver resection and liver transplantation. As excess accumulation of reactive oxygen species (ROS) is the leading factor, ceria nanoparticle, a cyclic reversible antioxidant, is an excellent candidate for HIRI. Methods Manganese doped mesoporous hollow ceria nanoparticles (MnOx-CeO2 NPs) were prepared, and the physicochemical characteristics, such as particle size, morphology, microstructure, etc. were elucidated. The in vivo safety and liver targeting effect were examined after i.v. injection. The anti-HIRI was determined by a mouse HIRI model. Results MnOx-CeO2 NPs with 0.40% Mn doped exhibited the strongest ROS-scavenging capability, which may due to the increased specific surface area and surface oxygen concentration. The nanoparticles accumulated in the liver after i.v. injection and exhibited good biocompatibility. In the HIRI mice model, MnOx-CeO2 NPs significantly reduced the serum ALT and AST level, decreased the MDA level and increased the SOD level in the liver, prevent pathological damages in the liver. Conclusion MnOx-CeO2 NPs were successfully prepared and it could significantly inhibit the HIRI after i.v. injection.
Collapse
Affiliation(s)
- Peiru Si
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jiaxing Lei
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Chen Yang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Peipei Zhang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Xiaojiao Li
- Biobank, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Shaohua Zheng
- Department of Anesthesiology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Qingqing Li
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Correspondence: Qingqing Li; Jiye Zhang, School of Pharmacy, Health Science Center, Xi’an Jiaotong University, No. 76 Yanta Westroad, Xi’an, People’s Republic of China, Email ;
| | - Jiye Zhang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| |
Collapse
|
43
|
Chao D, Dong Q, Yu Z, Qi D, Li M, Xu L, Liu L, Fang Y, Dong S. Specific Nanodrug for Diabetic Chronic Wounds Based on Antioxidase-Mimicking MOF-818 Nanozymes. J Am Chem Soc 2022; 144:23438-23447. [PMID: 36512736 DOI: 10.1021/jacs.2c09663] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chronic wound is a common complication for diabetic patients, which entails substantial inconvenience, persistent pain, and significant economic burden to patients. However, current clinical treatments for diabetic chronic wounds remain unsatisfactory. A prolonged but ineffective inflammation phase in chronic wounds is the primary difference between diabetic chronic wounds and normal wounds. Herein, we present an effective antioxidative system (MOF/Gel) for chronic wound healing of diabetic rats through integrating a metal organic framework (MOF) nanozyme with antioxidant enzyme-like activity with a hydrogel (Gel). MOF/Gel can continuously scavenge reactive oxygen species to modulate the oxidative stress microenvironment in diabetic chronic wounds, which leads to a natural transition from the inflammation phase to the proliferation phase. Impressively, the efficacy of one-time-applied MOF/Gel was comparable to that of the human epidermal growth factor Gel, a widely used clinical drug for various wound treatments. Such an effective, safe, and convenient MOF/Gel system can meet complex clinical demands.
Collapse
Affiliation(s)
- Daiyong Chao
- College of Chemistry, Jilin University, Changchun, Jilin 130012, China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Qing Dong
- College of Chemistry, Jilin University, Changchun, Jilin 130012, China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Zhixuan Yu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.,University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Desheng Qi
- College of Chemistry, Jilin University, Changchun, Jilin 130012, China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Minghua Li
- College of Chemistry, Jilin University, Changchun, Jilin 130012, China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Lili Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.,University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Ling Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Youxing Fang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Shaojun Dong
- College of Chemistry, Jilin University, Changchun, Jilin 130012, China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.,University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
44
|
Lu Y, Cao C, Pan X, Liu Y, Cui D. Structure design mechanisms and inflammatory disease applications of nanozymes. NANOSCALE 2022; 15:14-40. [PMID: 36472125 DOI: 10.1039/d2nr05276h] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Nanozymes are artificial enzymes with high catalytic activity, low cost, and good biocompatibility, and have received ever-increasing attention in recent years. Various inorganic and organic nanoparticles have been found to exhibit enzyme-like activities and are used as nanozymes for diverse biomedical applications ranging from tumor imaging and therapeutics to detection. However, their further clinical applications are hindered by the potential toxicity and long-term retention of nanomaterials in vivo. Clarifying the catalytic mechanism of nanozymes and identifying the key factors responsible for their behavior can guide the design of nanozyme structure, enlighten the ways to improve their enzyme-like activities, and minimize the dosage of nanozymes, leading to reduced toxicity to the human body for a real biomedical application prospect. In particular, inflammation occurring in numerous diseases is closely related to reactive oxygen species, and the active oxygen scavenging ability of nanozymes potentially exerts excellent therapeutic effects on inflammatory diseases. In this review, we systematically summarize the structure-activity relationship of nanozymes, including regulation strategies for size and morphology, surface structure, and composition. Based on the structure-activity mechanisms, a series of chemically designed nanozymes developed to target various inflammatory diseases are briefly summarized.
Collapse
Affiliation(s)
- Yi Lu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
| | - Cheng Cao
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
| | - Xinni Pan
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanlei Liu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
- National Engineering Center for Nanotechnology, Shanghai 200240, People's Republic of China.
| |
Collapse
|
45
|
|
46
|
Multi-enzyme activity nanozymes for biosensing and disease treatment. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
47
|
Meng L, Feng J, Gao J, Zhang Y, Mo W, Zhao X, Wei H, Guo H. Reactive Oxygen Species- and Cell-Free DNA-Scavenging Mn 3O 4 Nanozymes for Acute Kidney Injury Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50649-50663. [PMID: 36334088 DOI: 10.1021/acsami.2c16305] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Reactive oxygen species (ROS) scavenging therapy toward acute kidney injury (AKI) is promising, but no effective ROS scavenging drug has been developed yet. Moreover, cell-free DNA (cfDNA) is also involved in AKI, but the corresponding therapies have not been well developed. To tackle these challenges, Mn3O4 nanoflowers (Nfs) possessing both ROS and cfDNA scavenging activities were developed for better AKI protection as follows. First, Mn3O4 Nfs could protect HK2 cells through cascade ROS scavenging (dismutating ·O2- into H2O2 by superoxide dismutase-like activity and then decomposing H2O2 by catalase-like activity). Second, Mn3O4 Nfs could efficiently adsorb cfDNA and then decrease the inflammation caused by cfDNA. Combined, remarkable therapeutic efficacy was achieved in both cisplatin-induced and ischemia-reperfusion AKI murine models. Furthermore, Mn3O4 Nfs could be used for the T1-MRI real-time imaging of AKI. This study not only offered a promising treatment for AKI but also showed the translational potential of nanozymes.
Collapse
Affiliation(s)
- Longxiyu Meng
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology Nanjing University, Nanjing, Jiangsu 210008, China
| | - Jiayuan Feng
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Jie Gao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology Nanjing University, Nanjing, Jiangsu 210008, China
| | - Yihong Zhang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Wenjing Mo
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210008, China
| | - Xiaozhi Zhao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology Nanjing University, Nanjing, Jiangsu 210008, China
| | - Hui Wei
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210093, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu 210023 China
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology Nanjing University, Nanjing, Jiangsu 210008, China
| |
Collapse
|
48
|
Garrido MD, El Haskouri J, Marcos MD, Pérez-Pla F, Ros-Lis JV, Amorós P. One-Pot Synthesis of MnO x-SiO 2 Porous Composites as Nanozymes with ROS-Scavenging Properties. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3503. [PMID: 36234632 PMCID: PMC9565283 DOI: 10.3390/nano12193503] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
The development of nanomaterials that mimic the activity of enzymes is a topic of interest, for the decomposition of reactive oxygen species (ROS). We report the preparation of a novel nanocomposite of MnOx needles covered with SiO2 porous material. The material was prepared in one pot with a two-step procedure. The material was characterized by EDX, SEM, TEM, XRD, nitrogen adsorption-desorption isotherms, and XPS. The synthesis protocol took advantage of the atrane method, favoring the nucleation and initial growth of manganese oxide needles that remained embedded and homogeneously dispersed in a mesoporous silica matrix. The final composite had a high concentration of Mn (Si/Mn molar ratio of ca. 1). The nanozyme presented bimodal porosity: intraparticle and interparticle association with the surfactant micelles and the gaps between silica particles and MnOx needles, respectively. The porosity favored the migration of the reagent to the surface of the catalytic MnOx. The nanozyme showed very efficient SOD and catalase activities, thus improving other materials previously described. The kinetics were studied in detail, and the reaction mechanisms were proposed. It was shown that silica does not play an innocent role in the case of catalase activity, increasing the reaction rate.
Collapse
Affiliation(s)
- M. Dolores Garrido
- Institut de Ciència dels Materials (ICMUV), Universitat de València, Catedrático José Beltrán 2, 46980 Paterna, Spain
| | - Jamal El Haskouri
- Institut de Ciència dels Materials (ICMUV), Universitat de València, Catedrático José Beltrán 2, 46980 Paterna, Spain
| | - María D. Marcos
- Centro de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Unidad Mixta Universitat Politècnica de Valencia, Universitat de València, Departamento de Química, Universitat Politècnica de Valencia, 46022 Valencia, Spain
- CIBER de Bioingenieria, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Francisco Pérez-Pla
- Institut de Ciència dels Materials (ICMUV), Universitat de València, Catedrático José Beltrán 2, 46980 Paterna, Spain
| | - José Vicente Ros-Lis
- Centro de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Unidad Mixta Universitat Politècnica de Valencia, Universitat de València, Departamento de Química Inorgánica, Universitat de València, Doctor Moliner 56, 46100 Valencia, Spain
| | - Pedro Amorós
- Institut de Ciència dels Materials (ICMUV), Universitat de València, Catedrático José Beltrán 2, 46980 Paterna, Spain
| |
Collapse
|
49
|
Xu D, Wu L, Yao H, Zhao L. Catalase-Like Nanozymes: Classification, Catalytic Mechanisms, and Their Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203400. [PMID: 35971168 DOI: 10.1002/smll.202203400] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/08/2022] [Indexed: 06/15/2023]
Abstract
The field of nanozymes has developed rapidly over the past decade. Among various oxidoreductases mimics, catalase (CAT)-like nanozyme, acting as an essential part of the regulation of reactive oxygen species (ROS), has attracted extensive research interest in recent years. However, CAT-like nanozymes are not as well discussed as other nanozymes such as peroxidase (POD)-like nanozymes, etc. Compared with natural catalase or artificial CAT enzymes, CAT-like nanozymes have unique properties of low cost, size-dependent properties, high catalytic activity and stability, and easy surface modification, etc., which make them widely used in various fields, especially in tumor therapy and disease treatment. Consequently, there is a great requirement to make a systematic discussion on CAT-like nanozymes. In this review, some key aspects of CAT-like nanozymes are deeply summarized as: 1) Typical CAT-like nanozymes classified by different nanomaterials; 2) The catalytic mechanisms proposed by experimental and theoretical studies; 3) Extensive applications in regard to tumor therapy, cytoprotection and sensing. Therefore, it is prospected that this review will contribute to the further design of CAT-like nanozymes and optimize their applications with much higher efficiency than before.
Collapse
Affiliation(s)
- Deting Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Liyuan Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Haodong Yao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Lina Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
50
|
Hyaluronic acid-guided assembly of ceria nanozymes as plaque-targeting ROS scavengers for anti-atherosclerotic therapy. Carbohydr Polym 2022; 296:119940. [DOI: 10.1016/j.carbpol.2022.119940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 11/27/2022]
|