1
|
Pan J, Wang J, Wang W, Liu Z, Huo S, Yan L, Jiang W, Shao F, Gu Y. Renal-clearable and mitochondria-targeted metal-engineered carbon dot nanozymes for regulating mitochondrial oxidative stress in acute kidney injury. Mater Today Bio 2025; 32:101717. [PMID: 40242480 PMCID: PMC12002839 DOI: 10.1016/j.mtbio.2025.101717] [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: 01/09/2025] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Mitochondrial dysfunction-induced oxidative stress is a key pathogenic factor in acute kidney injury (AKI). Despite this, current mitochondrial-targeted antioxidant therapies have shown limited efficacy in clinical settings. In this study, we introduce a novel renal-clearable and mitochondria-targeted antioxidant nanozyme (TPP@RuCDzyme) designed to precisely modulate mitochondrial oxidative stress and mitigate AKI progression. TPP@RuCDzyme was synthesized by integrating ruthenium-doped carbon dots (CDs) with triphenylphosphine (TPP), a mitochondria-targeting moiety. This nanozyme system exhibits cascade enzyme-like activities, mimicking superoxide dismutase (SOD) and catalase (CAT), to efficiently convert cytotoxic superoxide (O2•-) and hydrogen peroxide (H2O2) into non-toxic water (H2O) and oxygen (O2). This dual-enzyme mimicry effectively alleviates mitochondrial oxidative damage, restores mitochondrial function, and inhibits apoptosis. Compared to RuCDzyme alone, TPP@RuCDzyme demonstrated significantly enhanced efficacy in alleviating glycerol-induced AKI by inhibiting oxidative stress. By leveraging the catalytic activity derived from the integration of CDs and a metallic element, this study presents a promising therapeutic strategy for AKI and other renal diseases associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jiangpeng Pan
- Department of Nephrology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, 450003, China
- Department of Nephrology, Henan Clinical Medical Research Center for Nephropathy, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital; Henan University People’s Hospital, Zhengzhou, Henan, 450003, China
| | - Juntao Wang
- Department of Nephrology, The First People's Hospital of Shangqiu, Shangqiu, Henan, China
| | - Wei Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
| | - Ziyang Liu
- Department of Nephrology, Henan Clinical Medical Research Center for Nephropathy, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital; Henan University People’s Hospital, Zhengzhou, Henan, 450003, China
| | - Shuai Huo
- Department of Nephrology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, 450003, China
| | - Lei Yan
- Department of Nephrology, Henan Clinical Medical Research Center for Nephropathy, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital; Henan University People’s Hospital, Zhengzhou, Henan, 450003, China
| | - Wei Jiang
- Department of Nephrology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, 450003, China
| | - Fengmin Shao
- Department of Nephrology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, 450003, China
- Department of Nephrology, Henan Clinical Medical Research Center for Nephropathy, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital; Henan University People’s Hospital, Zhengzhou, Henan, 450003, China
| | - Yue Gu
- Department of Nephrology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, 450003, China
- Department of Nephrology, Henan Clinical Medical Research Center for Nephropathy, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital; Henan University People’s Hospital, Zhengzhou, Henan, 450003, China
| |
Collapse
|
2
|
Wan L, Li S, Du J, Li A, Zhan Y, Zhu W, Zheng P, Qiao D, Nie C, Pan Q. Review of Metal-Polyphenol Self-Assembled Nanoparticles: Synthesis, Properties, and Biological Applications in Inflammatory Diseases. ACS Biomater Sci Eng 2025. [PMID: 40276988 DOI: 10.1021/acsbiomaterials.4c02366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Polyphenols, which are compounds characterized by the presence of phenolic hydroxyl groups, are abundantly found in natural plants and exist in highly complex forms within living organisms. As some of the most prevalent compounds in nature, polyphenols possess significant medicinal value due to their unique structural features, particularly their therapeutic efficacy in antitumor, anti-inflammatory, and antibacterial applications. In the context of inflammation therapy, polyphenolic compounds can inhibit the excessive release of inflammatory mediators from inflammatory cells, thereby mitigating inflammation. Furthermore, these compounds exhibit strong antioxidant properties, enabling them to scavenge free radicals and reactive oxygen species (ROS), reduce oxidative stress-related damage, and exert anti-inflammatory effects. Due to their multiple phenolic hydroxyl groups and their ability to chelate various metals, polyphenols are extensively utilized in the synthesis of self-assembled nanoparticles for the treatment of various diseases. Numerous studies have demonstrated that the therapeutic profile of nanoparticles formed through self-assembly with metal ions surpasses that of polyphenolic compounds alone. This Review will focus on the self-assembly of different polyphenolic compounds with various metal ions to generate nanoparticles, their characterization, and their therapeutic applications in inflammation-related diseases, providing researchers with new insights into the synthetic study of metal-polyphenol nanocomposites and their biological applications.
Collapse
Affiliation(s)
- Li Wan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Shizhe Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Jiawei Du
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Anqi Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Yujie Zhan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Pengwu Zheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Dan Qiao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Cunpeng Nie
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Qingshan Pan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| |
Collapse
|
3
|
Luo A, Tan J, Wang H, Yang M, Wang S, Wang C, Wang Y, Liu L. Engineered Prussian Blue-Curcumin Nanozyme with RONS Scavenging Properties for Augmented Reversible Treatment of Cardiac Hypertrophy. Adv Healthc Mater 2025:e2404201. [PMID: 40270221 DOI: 10.1002/adhm.202404201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/28/2025] [Indexed: 04/25/2025]
Abstract
Pathological cardiac hypertrophy, often triggered by the excessive production and accumulation of reactive oxygen and nitrogen species (RONS), may ultimately lead to heart failure. The treatment of myocardial hypertrophy often involves antioxidant stress therapy. In this study, by coordinating curcumin with ferric ions during the synthesis of Prussian blue nanoparticles, a Prussian blue-curcumin (PB-Cur) nanozyme is successfully engineered with exceptional reactive oxygen and nitrogen species (RONS) elimination capabilities. Following PVP modification, the PB-Cur nanozyme exhibited favorable biocompatibility and stability in aqueous solutions. Furthermore, the PB-Cur nanozyme shows remarkable reversible treatment efficacy against myocardial hypertrophy in both in vitro and in vivo models. After one week of treatment, the PB-Cur group in the transverse aortic constriction (TAC)-induced cardiac hypertrophy models displayed a notable decrease in myocardial hypertrophy and fibrosis. Echocardiographic findings also revealed a substantial improvement in cardiac function among TAC mice following PB-Cur administration. Mechanistically, through reactive oxygen species (ROS) elimination, the PB-Cur effectively downregulated oxidative stress-related pathways, including MAPK and PI3K-Akt, which hold promise for treating oxidative stress-related cardiac diseases.
Collapse
Affiliation(s)
- Anlin Luo
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
- Henan Institute of Interconnected Intelligent Health Management, Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, 450052, China
| | - Jing Tan
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
- Henan Institute of Interconnected Intelligent Health Management, Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, 450052, China
| | - Haisong Wang
- Reproductive Medical Center, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Manman Yang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
- Henan Institute of Interconnected Intelligent Health Management, Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, 450052, China
| | - Shuojing Wang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
- Henan Institute of Interconnected Intelligent Health Management, Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, 450052, China
| | - Chengzeng Wang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
- Henan Institute of Interconnected Intelligent Health Management, Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, 450052, China
| | - Yachao Wang
- Henan Institute of Interconnected Intelligent Health Management, Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, 450052, China
| | - Lin Liu
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
- Henan Institute of Interconnected Intelligent Health Management, Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, Henan, 450052, China
| |
Collapse
|
4
|
Yang H, Zhang X, Wu J, Xiao Y, Dai L, Wang G, Zhang X, Hu C, He S, Yuan Z. Probiotic Membrane-Modified Nanocomposite Alleviates Inflammation and Microbiota Dysbiosis in Colitis by Scavenging Oxidative Stress and Restoring Immune Homeostasis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22245-22265. [PMID: 40184333 DOI: 10.1021/acsami.4c22004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2025]
Abstract
Inflammatory bowel disease (IBD) is a complex chronic intestinal disorder in which excessive oxidative stress, dysregulated immune response, and microbiota dysbiosis contribute to recurrent episodes of inflammation in the colonic mucosa. Current clinical treatments focusing solely on inflammation resolution often exhibit limited efficacy due to the inability to fundamentally improve the pathological microenvironment. Herein, a probiotic membrane-modified drug delivery nanocomposite, namely, MPDA@Cur@EM, is developed for the comprehensive treatment of IBD. It contains two components: the curcumin-loaded mesoporous polydopamine nanoparticle (MPDA@Cur) as the core and the Escherichia coli Nissle 1917 outer membrane (EM) as the surface. For MPDA@Cur, the pathological microenvironment triggers the responsive release of curcumin. Importantly, MPDA@Cur can effectively alleviate the inflammatory response of LPS-activated macrophages through MPDA-mediated ROS scavenging and curcumin-induced M2 polarization. In the dextran sulfate sodium (DSS)-induced colitis model, the EM coating not only allows for the targeting enrichment of orally administered MPDA@Cur@EM to the inflamed colonic mucosa, but also participates in the regulation of intestinal flora. Consequently, MPDA@Cur@EM efficiently attenuates the inflammatory reaction and restores the intestinal barrier functions, demonstrated by the multipronged manner of restoring redox balance, remodeling immune homeostasis, and reshaping the gut microecology. Collectively, this work provides a safe and promising codelivery strategy of probiotic product, antioxidative nanoenzyme, and therapeutic drug for comprehensive management of IBD.
Collapse
Affiliation(s)
- Huan Yang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Xu Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Jianshuang Wu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Yao Xiao
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Liangliang Dai
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Gaoyang Wang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Xiaohong Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Chenghu Hu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Shuixiang He
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Zhang Yuan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| |
Collapse
|
5
|
Yang M, Deng Z, Zhu Y, Xu C, Ding C, Zhang Y, Zhang M, Zhang M. Advancements in herbal medicine-based nanozymes for biomedical applications. Chin Med J (Engl) 2025:00029330-990000000-01500. [PMID: 40169370 DOI: 10.1097/cm9.0000000000003584] [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: 09/24/2024] [Indexed: 04/03/2025] Open
Abstract
ABSTRACT Nanozymes are a distinct category of nanomaterials that exhibit catalytic properties resembling those of enzymes such as peroxidase (POD), superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). Nanozymes derived from Chinese herbal medicines exhibit the catalytic functions of their enzyme mimics, while retaining the specific medicinal properties of the herb (termed "herbzymes"). These nanozymes can be categorized into three main groups based on their method of synthesis: herb carbon dot nanozymes, polyphenol-metal nanozymes, and herb extract nanozymes. The reported catalytic activities of herbzymes include POD, SOD, CAT, and GPx. This review presents an overview of the catalytic activities and potential applications of nanozymes, introduces the novel concept of herbzymes, provides a comprehensive review of their classification and synthesis, and discusses recent advances in their biomedical applications. Furthermore, we also discuss the significance of research into herbzymes, including the primary challenges faced and future development directions.
Collapse
Affiliation(s)
- Mei Yang
- Department of Organ Procurement and Allocation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhichao Deng
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yuanyuan Zhu
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chenxi Xu
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chenguang Ding
- Department of Organ Procurement and Allocation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Department of Kidney Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Mingxin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China
| |
Collapse
|
6
|
Zang Y, Zhang W, Wang P, Zhu C, Guo X, Wang W, Cheng L, Chen XL, Wang X. Bi 2Se 3/PAAS Hydrogels with Photothermal and Antioxidant Properties for Bacterial Infection Wound Therapy by Improving Vascular Function and Regulating Glycolipid Metabolism. Adv Healthc Mater 2025; 14:e2401810. [PMID: 39180451 DOI: 10.1002/adhm.202401810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/09/2024] [Indexed: 08/26/2024]
Abstract
Skin is the largest organ in the human body, and it is also the most important natural barrier. However, some accidents can cause skin damage. Bacterial infections and inflammatory reactions can hinder wound healing. Therefore, eliminating bacterial infections and regulating oxidative stress are essential. The use of antibiotics is no longer sufficient because of bacterial resistance. The development of new nanomaterials provides another way of thinking about bacterial drug resistance. In this study, bismuth selenide is modified with polyethylpyrrolidone to obtain a 2D nanomaterial with negligible toxicity and then added to a sodium polyacrylate hydrogel, which is nontoxic and has strong tissue adhesion and a weak antibacterial effect. To further enhance antibacterial performance, photothermal therapy is a good strategy. Under near-infrared light, Bi2Se3/PAAS shows a strong bactericidal effect. Bi2Se3/PAAS hydrogels also have certain antioxidant effects and are used to remove excess free radicals from wound infections. The effective therapeutic effect of Bi2Se3/PAAS/NIR on methicillin-resistant Staphylococcus aureus (MRSA) infection is further verified in animal models. Transcriptome analysis reveals that the Bi2Se3/PAAS hydrogel improves the function of vascular endothelial cells, regulates glucose and lipid metabolism, and promotes the healing of infected wounds.
Collapse
Affiliation(s)
- Ying Zang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, P. R. China
| | - Wei Zhang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Peisan Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Can Zhu
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Xueting Guo
- School of Pharmacy, Anhui Medical University, Hefei, 230032, P. R. China
| | - Wenqi Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, P. R. China
| | - Xianwen Wang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, P. R. China
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| |
Collapse
|
7
|
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
|
8
|
Zhang R, Liu Y, Gao Y, Peng D, Luan Q, Li Z, Xia X, Xiang X. Flavonoid-rich sesame leaf extract-mediated synthesis of nanozymes: Extraction optimization, chemical composition identification and bioactivity evaluation. Food Chem 2024; 456:140021. [PMID: 38870817 DOI: 10.1016/j.foodchem.2024.140021] [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: 03/19/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Sesame leaves contain rich phenolic acids and flavonoids. However, their potential in nanozyme synthesis has not been investigated yet. Herein, we report the preparation of flavonoid-rich sesame leaf extract (SLE), composition identification, and its use in the construction of iron (Fe)-based nanozymes (Fe-SLE CPNs). SLE was obtained with an extraction yield of ∼14.5% with a total flavonoid content (TFC) of ∼850.85 mg RE/g. There were 83 flavonoid compounds in SLE, primarily including scutellarin, apigenin-7-glucuronid, narcissin, and hyperoside. Fe-SLE CPNs exhibited nanodot morphology with a hydrodynamic size of 79.34 nm and good stability in various physiological solutions, pH levels, and temperatures. The Fe-SLE CPNs were more efficient in the scavenging ability of reactive oxygen species (ROS) than SLE alone. Furthermore, a stronger anti-inflammatory effect of the Fe-SLE CPNs was shown by modulating the MyD88-NF-κB-MAPK signaling pathways. These findings imply that SLE-based nanozymes hold great potential for diverse applications.
Collapse
Affiliation(s)
- Ruiying Zhang
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China; School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Yufei Liu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Yiqiao Gao
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Dengfeng Peng
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China
| | - Qian Luan
- Ningbo Innovation Center, Zhejiang University, Ningbo 315100, China
| | - Ziliang Li
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China; School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Xiaoyang Xia
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China.
| | - Xia Xiang
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China.
| |
Collapse
|
9
|
Liu X, Guo Y, Pan J, Wu T, Zhao B, Wei S, Jiang W, Liu Y. Nanoparticles constructed from natural polyphenols are used in acute kidney injury. J Mater Chem B 2024; 12:8883-8896. [PMID: 39177039 DOI: 10.1039/d4tb00837e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Acute kidney injury (AKI) is a severe clinical syndrome characterized by rapid deterioration of renal function caused by a variety of pathogeneses. Natural polyphenols have been considered to have potential in the treatment of AKI due to their powerful antioxidant and anti-inflammatory activities, but their low bioavailability in vivo limits their efficacy. Polyphenol nanoparticles based on a nano-delivery system show good effects in reducing kidney injury, improving renal function and promoting renal tissue repair, and brings new hope and possibility for the treatment of AKI. This review provides an overview of the common characteristics, treatments, and associated adverse effects of AKI. The classification and bioavailability of polyphenols as well as their therapeutic role in AKI and potential possible effects are outlined. The potential therapeutic effects of polyphenol-based nanoparticles on AKI and the underlying mechanisms are discussed.
Collapse
Affiliation(s)
- Xiaohua Liu
- Henan Science and Technology Innovation Promotion Center, Zhengzhou 450046, China
| | - Yike Guo
- Department of Pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jiangpeng Pan
- Department of Pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Tingting Wu
- Department of Pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| | - Bing Zhao
- Henan Finance University, Zhengzhou 450046, China
| | - Shuyi Wei
- Plastic Surgery Department, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, China.
| | - Wei Jiang
- Department of Pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Liu
- Department of Pharmacy, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China.
| |
Collapse
|
10
|
Fang F, Chen X. Carrier-Free Nanodrugs: From Bench to Bedside. ACS NANO 2024; 18:23827-23841. [PMID: 39163559 DOI: 10.1021/acsnano.4c09027] [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: 08/22/2024]
Abstract
Carrier-free nanodrugs with extraordinary active pharmaceutical ingredient (API) loading (even 100%), avoidable carrier-induced toxicity, and simple synthetic procedures are considered as one of the most promising candidates for disease theranostics. Substantial studies and the commercial success of "carrier-free" nanocrystals have demonstrated their strong clinical potential. However, their practical translations remain challenging and are impeded by unpredictable assembly processes, insufficient delivery efficiency, and an unclear in vivo fate. In this Perspective, we systematically outline the contemporary and emerging carrier-free nanodrugs based on diverse APIs, as well as highlight their opportunities and challenges in clinical translation. Looking ahead, further improvements in design and preparation, drug delivery, in vivo efficacy, and safety of carrier-free nanomedicines are essential to facilitate their translation from the bench to bedside.
Collapse
Affiliation(s)
- Fang Fang
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| |
Collapse
|
11
|
Yao H, Zhu Z, Liu M, Sun F, Du M, Sun Y, Du B. Multifunctional Nanosystem Based on Ultrasmall Carbon Dots for the Treatment of Acute Kidney Injury. ACS Biomater Sci Eng 2024; 10:4970-4984. [PMID: 39022808 DOI: 10.1021/acsbiomaterials.4c00616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Acute kidney injury (AKI) is a critical medical condition characterized by high morbidity and mortality rates. The pathogenesis of AKI potentially involves bursts of reactive oxygen species (ROS) bursts and elevated levels of inflammatory mediators. Developing nanoparticles (NPs) that downregulate ROS and inflammatory mediators is a promising approach to treat AKI. However, such NPs would be affected by the glomerular filtration barrier (GFB). Typically, NPs are too large to penetrate the glomerular system and reach the renal tubules─the primary site of AKI injury. Herein, we report the development of ultrasmall carbon dots-gallic acid (CDs-GA) NPs (∼5 nm). These NPs exhibited outstanding biocompatibility and were shown not only to efficiently eliminate ROS and alleviate oxidative stress but also to suppress the activation of the NF-κB signaling pathway, leading to a reduction in the release of inflammatory factors. Importantly, CDs-GA NPs were shown to be able to rapidly accumulate rapidly in the renal tissues without the need for intricate targeting strategies. In vivo studies demonstrated that CDs-GA NPs significantly reduced the incidence of cisplatin (CDDP)-induced AKI in mice, surpassing the efficacy of the small molecular drug, N-acetylcysteine. This research provides an innovative strategy for the treatment of AKI.
Collapse
Affiliation(s)
- Hanchun Yao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Zhihui Zhu
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Mengyu Liu
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Fangfang Sun
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Mengyu Du
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Yilin Sun
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Du
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| |
Collapse
|
12
|
Cao F, Liang K, Tang WW, Ni QY, Ji ZY, Zha CK, Wang YK, Jiang ZX, Hou S, Tao LM, Wang X. Polyvinylpyrrolidone-curcumin nanoparticles with immune regulatory and metabolism regulatory effects for the treatment of experimental autoimmune uveitis. J Control Release 2024; 372:551-570. [PMID: 38914206 DOI: 10.1016/j.jconrel.2024.06.047] [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: 02/28/2024] [Revised: 05/30/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
Uveitis comprises a cluster of intraocular inflammatory disorders characterized by uncontrolled autoimmune responses and excessive oxidative stress leading to vision loss worldwide. In the present study, curcumin (CUR) was conjugated with polyvinylpyrrolidone (PVP) to form PVP-CUR nanoparticles with significantly elevated solubility and outstanding multiple radical scavenging abilities. In vitro studies revealed that PVP-CUR nanoparticles markedly mitigated oxidative stress and reduced apoptosis in a H2O2-induced human retinal pigment epithelial cell line (ARPE-19) and promoted phenotypic polarization from M1 to M2 in an LPS-induced human microglial cell line (HMC3). Further in vivo studies demonstrated the prominent therapeutic effects of PVP-CUR nanoparticles on experimental autoimmune uveitis (EAU), which relieved clinical and pathological progression, improved perfusion and tomographic manifestations of retinal vessels, and reduced blood-retinal barrier (BRB) leakage; these effects may be mediated by mitigating oxidative stress and attenuating macrophage/microglia-elicited inflammation. Notably, treatment with PVP-CUR nanoparticles was shown to regulate metabolite alterations in EAU rats, providing novel insights into the underlying mechanisms involved. Additionally, the PVP-CUR nanoparticles showed great biocompatibility in vivo. In summary, our study revealed that PVP-CUR nanoparticles may serve as effective and safe nanodrugs for treating uveitis and other oxidative stress- and inflammation-related diseases.
Collapse
Affiliation(s)
- Fan Cao
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Clinical Medicine, The Second School of Clinical Medicine, Anhui Medical University, Hefei 230032, PR China
| | - Kun Liang
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Wei-Wei Tang
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Qin-Yu Ni
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Clinical Medicine, The Second School of Clinical Medicine, Anhui Medical University, Hefei 230032, PR China
| | - Zhi-Yu Ji
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Clinical Medicine, The Second School of Clinical Medicine, Anhui Medical University, Hefei 230032, PR China
| | - Chen-Kai Zha
- Department of Clinical Medicine, The Second School of Clinical Medicine, Anhui Medical University, Hefei 230032, PR China
| | - Ya-Kun Wang
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Zheng-Xuan Jiang
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Shengping Hou
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, PR China.
| | - Li-Ming Tao
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Xianwen Wang
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, PR China.
| |
Collapse
|
13
|
Pereira GC. A novel degradable PEG superparamagnetic iron oxide capsule coupled with a polyphenolic nano-enzymatic conjugate (PSPM-NE), to treat ROS-driven cardiovascular-diseases, tested in atherosclerosis as a model disease, and hypothesizing autoimmunity as an atheroma's trigger. Front Cardiovasc Med 2024; 11:1125571. [PMID: 39145281 PMCID: PMC11323396 DOI: 10.3389/fcvm.2024.1125571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/16/2024] [Indexed: 08/16/2024] Open
Abstract
Cardiovascular diseases account for a significant portion of the worldwide mortality rate. This aroused interest among the specialised scientific community, seeking for solutions based on non-clinical and clinical investigations, to shed light onto the physio-pathology of cardiovascular impairment. It is proven challenging managing chronic cardiovascular illnesses like atherosclerosis, arrhythmias, and diverse cardiomyopathies. In certain cases, there is no approved treatment. In other cases, the need for combining therapeutic components, when dealing with co-morbidities, may increase the risk of toxicity-driven cardiovascular impairment. In this case, because the risk of cardiac events correlates with the QT prolongation rates, the QT or QTc interval prolongation has become an important biomarker to access drug-related cardio-toxicity. Several approaches have been found in the current literature, aiming at improving physiological acceptance, i.e., to reduce toxicity. Nanotechnology has increasingly appeared as a promising ally to modulate active substances, preserving cardiovascular function and optimising drug effectiveness, i.e., acting as a cardio-protective mechanism, leveraging the effects of drug-driven cardio-toxicity. In this manuscript, the author combines plant active compounds and nanotechnological strategies, e.g., nano-encapsulation, nano-enzymes, magnetically driven nano-delivery systems, applied in regenerative medicine, and assesses their effects on the cardiovascular system, e.g., as cardio-protective factors, reducing cardio-toxicity. The aim is to propose a new strategy to tackle atherosclerosis initiation and progression, in a drug design that targets ROS-removal and reduces inflammation, using auto-immunity biomarkers to select key atheroma-related signalling cascades. To analyse physiological phenomena related to atherosclerosis initiation and progression, the author proposes both experimental observations and a new haemorheological computational model of arterial constriction. The results of such analysis are used as motivators in the design of the here presented strategy to tackle atheroma. This novel design is based on degradable polyethylene glycol (PEG) superparamagnetic iron oxide capsule coupled with a polyphenolic nano-enzymatic conjugate (PSPM-NE).
Collapse
Affiliation(s)
- Glaucia C. Pereira
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London, United Kingdom
| |
Collapse
|
14
|
Shan J, Jin X, Zhang C, Huang M, Xing J, Li Q, Cui Y, Niu Q, Chen XL, Wang X. Metal natural product complex Ru-procyanidins with quadruple enzymatic activity combat infections from drug-resistant bacteria. Acta Pharm Sin B 2024; 14:2298-2316. [PMID: 38799629 PMCID: PMC11121202 DOI: 10.1016/j.apsb.2023.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 05/29/2024] Open
Abstract
Bacterial infection hampers wound repair by impeding the healing process. Concurrently, inflammation at the wound site triggers the production of reactive oxygen species (ROS), causing oxidative stress and damage to proteins and cells. This can lead to chronic wounds, posing severe risks. Therefore, eliminating bacterial infection and reducing ROS levels are crucial for effective wound healing. Nanozymes, possessing enzyme-like catalytic activity, can convert endogenous substances into highly toxic substances, such as ROS, to combat bacteria and biofilms without inducing drug resistance. However, the current nanozyme model with single enzyme activity falls short of meeting the complex requirements of antimicrobial therapy. Thus, developing nanozymes with multiple enzymatic activities is essential. Herein, we engineered a novel metalloenzyme called Ru-procyanidin nanoparticles (Ru-PC NPs) with diverse enzymatic activities to aid wound healing and combat bacterial infections. Under acidic conditions, due to their glutathione (GSH) depletion and peroxidase (POD)-like activity, Ru-PC NPs combined with H2O2 exhibit excellent antibacterial effects. However, in a neutral environment, the Ru-PC NPs, with catalase (CAT) activity, decompose H2O2 to O2, alleviating hypoxia and ensuring a sufficient oxygen supply. Furthermore, Ru-PC NPs possess exceptional antioxidant capacity through their superior superoxide dismutase (SOD) enzyme activity, effectively scavenging excess ROS and reactive nitrogen species (RNS) in a neutral environment. This maintains the balance of the antioxidant system and prevents inflammation. Ru-PC NPs also promote the polarization of macrophages from M1 to M2, facilitating wound healing. More importantly, Ru-PC NPs show good biosafety with negligible toxicity. In vivo wound infection models have confirmed the efficacy of Ru-PC NPs in inhibiting bacterial infection and promoting wound healing. The focus of this work highlights the quadruple enzymatic activity of Ru-PC NPs and its potential to reduce inflammation and promote bacteria-infected wound healing.
Collapse
Affiliation(s)
- Jie Shan
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Xu Jin
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Cong Zhang
- Division of Gastroenterology, the First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Muchen Huang
- The Second Clinical Medical College, Anhui Medical University, Hefei 230022, China
| | - Jianghao Xing
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qingrong Li
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Yuyu Cui
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qiang Niu
- The Second Clinical Medical College, Anhui Medical University, Hefei 230022, China
| | - Xu Lin Chen
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
- College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| |
Collapse
|
15
|
Sun J, Zhao X, Shen H, Dong J, Rong S, Cai W, Zhang R. CD44-targeted melanin-based nanoplatform for alleviation of ischemia/reperfusion-induced acute kidney injury. J Control Release 2024; 368:1-14. [PMID: 38367863 DOI: 10.1016/j.jconrel.2024.02.021] [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: 09/25/2023] [Revised: 01/24/2024] [Accepted: 02/14/2024] [Indexed: 02/19/2024]
Abstract
Ischemia/reperfusion (I/R)-induced acute kidney injury (AKI) is a serious kidney disease with high morbidity and mortality. However, there is no effective clinical treatment strategy. Herein, we developed a CD44 targeting nanoplatform based on HA-assembled melanin NPs covalently coupled with dexamethasone for I/R-induced AKI therapy by alleviating oxidative/inflammatory- induced damage. The constructed HA-MNP-DXM NPs had good dispersion, stability, and broad-spectrum scavenging capabilities against multiple reactive free radicals. Moreover, the NPs could be efficiently internalized and exhibited antioxidative, anti-inflammatory, and antiapoptotic effects in CoCl2-stimulated renal tubular epithelial NRK-52E cells. Furthermore, the I/R-induced AKI murine model was established to evaluate the in vivo performance of NPs. The results suggested the NPs could specifically target impaired kidneys upon intravenous administration according to NIR-II fluorescence imaging and showed high biosafety. Importantly, the NPs could improve renal function, alleviate oxidative stress and inflammatory reactions, inhibit apoptosis of tubular cells, and restore mitochondrial structure and function, exhibiting excellent therapeutic effects. Further therapeutic mechanism indicated the NPs maintained the cellular/mitochondrial redox balance by modulating the Nrf2 and HO-1 expression. Therefore, the NPs can be a promising therapeutic candidate for the treatment of I/R-induced AKI.
Collapse
Affiliation(s)
- Jinghua Sun
- First Hospital of Shanxi Medical University, Taiyuan, 030001, China; Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xuhui Zhao
- First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Hao Shen
- School of Pharmacy, Shanxi Medical University, Taiyuan, 030001, China
| | - Jie Dong
- Shanxi Medical University, Taiyuan, 030001, China
| | - Shuo Rong
- Shanxi Medical University, Taiyuan, 030001, China
| | - Wenwen Cai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People' Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
16
|
Zhang R, Thoröe-Boveleth S, Chigrin DN, Kiessling F, Lammers T, Pallares RM. Nanoscale engineering of gold nanostars for enhanced photoacoustic imaging. J Nanobiotechnology 2024; 22:115. [PMID: 38493118 PMCID: PMC10943878 DOI: 10.1186/s12951-024-02379-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/28/2024] [Indexed: 03/18/2024] Open
Abstract
Photoacoustic (PA) imaging is a diagnostic modality that combines the high contrast resolution of optical imaging with the high tissue penetration of ultrasound. While certain endogenous chromophores can be visualized via PA imaging, many diagnostic assessments require the administration of external probes. Anisotropic gold nanoparticles are particularly valued as contrast agents, since they produce strong PA signals and do not photobleach. However, the synthesis of anisotropic nanoparticles typically requires cytotoxic reagents, which can hinder their biological application. In this work, we developed new PA probes based on nanostar cores and polymeric shells. These AuNS were obtained through one-pot synthesis with biocompatible Good's buffers, and were subsequently functionalized with polyethylene glycol, chitosan or melanin, three coatings widely used in (pre)clinical research. Notably, the structural features of the nanostar cores strongly affected the PA signal. For instance, despite displaying similar sizes (i.e. 45 nm), AuNS obtained with MOPS buffer generated between 2 and 3-fold greater signal intensities in the region between 700 and 800 nm than nanostars obtained with HEPES and EPPS buffers, and up to 25-fold stronger signals than spherical gold nanoparticles. A point source analytical model demonstrated that AuNS synthesized with MOPS displayed greater absorption coefficients than the other particles, corroborating the stronger PA responses. Furthermore, the AuNS shell not only improved the biocompatibility of the nanoconstructs but also affected their performance, with melanin coating enhancing the signal more than 4-fold, due to its own PA capacity, as demonstrated by both in vitro and ex vivo imaging. Taken together, these results highlight the strengths of gold nanoconstructs as PA probes and offer insights into the design rules for the nanoengineering of new nanodiagnostic agents.
Collapse
Affiliation(s)
- Rui Zhang
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Sven Thoröe-Boveleth
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany
| | - Dmitry N Chigrin
- Institute of Physics (1A), RWTH Aachen University, 52056, Aachen, Germany
- DWI - Leibniz Institute for Interactive Materials, 52076, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Roger M Pallares
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074, Aachen, Germany.
| |
Collapse
|
17
|
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
|
18
|
Yu B, Jin Q, Ji J. Natural products applied in acute kidney injury treatment: polymer matters. Biomater Sci 2024; 12:621-633. [PMID: 38131274 DOI: 10.1039/d3bm01772a] [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: 12/23/2023]
Abstract
Acute kidney injury (AKI) is a global health threat due to its high morbidity and mortality. There is still a lack of effective therapeutic methods to deal with AKI clinically. Natural products with outstanding accessibility and bioactivity are potential candidates for AKI treatment. Natural product-based prodrugs or nano-structures with improved properties are frequently fabricated for maximizing bioavailability and decreasing side effects, in which natural polymers are selected as carriers, or natural drugs are loaded as cargos on designed polymers. In this review, the etiologies of AKI are briefly presented, and emerging natural products delivered rationally for AKI therapy, as either carriers or cargos, are both introduced. Moreover, the challenges of the future development of nature-based nanodrugs or prodrugs for AKI have also been discussed.
Collapse
Affiliation(s)
- Bo Yu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Qiao Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
19
|
Yang P, Huang Q, Zhang J, Li Y, Gao H, Gu Z. Natural Polyphenolic Nanodots for Alzheimer's Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308393. [PMID: 38010256 DOI: 10.1002/adma.202308393] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/07/2023] [Indexed: 11/29/2023]
Abstract
The abnormal amyloid-β accumulation is essential and obbligato in Alzheimer's disease pathogenesis and natural polyphenols exhibit great potential as amyloid aggregation inhibitors. However, the poor metabolic stability, low bioavailability, and weak blood-brain barrier crossing ability of natural polyphenol molecules fail to meet clinical needs. Here, a universal protocol to prepare natural polyphenolic nanodots is developed by heating in aqueous solution without unacceptable additives. The nanodots are able to not only inhibit amyloid-β fibrillization and trigger the fibril disaggregation, but mitigate the amyloid-β-plaque-induced cascade impairments including normalizing oxidative microenvironment, altering microglial polarization, and rescuing neuronal death and synaptic loss, which results in significant improvements in recognition and cognition deficits in transgenic mice. More importantly, natural polyphenolic nanodots possess stronger antiamyloidogenic performance compared with small molecule, as well as penetrate the blood-brain barrier. The excellent biocompatibility further guarantees the potential of natural polyphenolic nanodots for clinical applications. It is expected that natural polyphenolic nanodots provide an attractive paradigm to support the development of the therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Peng Yang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- Department of Health Products Technical Research and Development Center, Yunnanbaiyao Group Co. Ltd., Kunming, 650500, China
| | - Qianqian Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Jianhua Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Yiwen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Zhipeng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Lu Y, Pan X, Cao C, Fan S, Tan H, Cui S, Liu Y, Cui D. MnO 2 Coated Mesoporous PdPt Nanoprobes for Scavenging Reactive Oxygen Species and Solving Acetaminophen-Induced Liver Injury. Adv Healthc Mater 2023; 12:e2300163. [PMID: 37184887 DOI: 10.1002/adhm.202300163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/23/2023] [Indexed: 05/16/2023]
Abstract
As one of the most widely used drugs, acetaminophen, is the leading cause of acute liver injury. In addition, acetaminophen-induced liver injury (AILI) has a strong relationship with the overproduced reactive oxygen species, which can be effectively eliminated by nanozymes. To address these challenges, mesoporous PdPt@MnO2 nanoprobes (PPM NPs) mimicking peroxide, catalase, and superoxide dismutase-like properties are synthesized. They demonstrate nontoxicity, high colloidal stability, and exceptional reactive oxygen species (ROS)-scavenging ability. By scavenging excessive ROS, decreasing inflammatory cytokines, and inhibiting the recruitment and activation of monocyte/macrophage cells and neutrophils, the pathology mechanism of PPM NPs in AILI is confirmed. Moreover, PPM NPs' therapeutic effect and good biocompatibility may facilitate the clinical treatment of AILI.
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, P. R. China
| | - Xinni Pan
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200235, P. R. 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, P. R. China
| | - Shanshan Fan
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200235, P. R. China
| | - Haisong Tan
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Shengsheng 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, P. R. 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, P. R. 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, P. R. China
| |
Collapse
|
23
|
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
|
24
|
Ma H, Zhang X, Liu L, Huang Y, Sun S, Chen K, Xin Q, Liu P, Yan Y, Wang Y, Li Y, Liu H, Zhao R, Tan K, Chen X, Yuan X, Li Y, Liu Y, Dai H, Liu C, Wang H, Zhang XD. Bioactive NIR-II gold clusters for three-dimensional imaging and acute inflammation inhibition. SCIENCE ADVANCES 2023; 9:eadh7828. [PMID: 37531420 PMCID: PMC10396295 DOI: 10.1126/sciadv.adh7828] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/03/2023] [Indexed: 08/04/2023]
Abstract
Strong fluorescence and high catalytic activities cannot be achieved simultaneously due to conflicts in free electron utilization, resulting in a lack of bioactivity of most near-infrared-II (NIR-II) fluorophores. To circumvent this challenge, we developed atomically precise Au22 clusters with strong NIR-II fluorescence ranging from 950 to 1300 nm exhibiting potent enzyme-mimetic activities through atomic engineering to create active Cu single-atom sites. The developed Au21Cu1 clusters show 18-fold higher antioxidant, 90-fold higher catalase-like, and 3-fold higher superoxide dismutase-like activities than Au22 clusters, with negligible fluorescence loss. Doping with single Cu atoms decreases the bandgap from 1.33 to 1.28 eV by predominant contributions from Cu d states, and Cu with lost electron states effectuates high catalytic activities. The renal clearable clusters can monitor cisplatin-induced renal injury in the 20- to 120-minute window and visualize it in three dimensions using NIR-II light-sheet microscopy. Furthermore, the clusters inhibit oxidative stress and inflammation in the cisplatin-treated mouse model, particularly in the kidneys and brain.
Collapse
Affiliation(s)
- Huizhen Ma
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Xiaoning Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Ling Liu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - You Huang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Si Sun
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Ke Chen
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Qi Xin
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Pengfei Liu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Yuxing Yan
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Yili Wang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Yuan Li
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Haile Liu
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Ruoli Zhao
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Kexin Tan
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Xinzhu Chen
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Xun Yuan
- School of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao, Shandong 266042, China
| | - Yonghui Li
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Ying Liu
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Haitao Dai
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Changlong Liu
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Hao Wang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Xiao-Dong Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China
| |
Collapse
|
25
|
Luo X, Xiong H, Jiang Y, Fan Y, Zuo C, Chen D, Chen L, Lin H, Gao J. Macrophage Reprogramming via Targeted ROS Scavenging and COX-2 Downregulation for Alleviating Inflammation. Bioconjug Chem 2023. [PMID: 37330989 DOI: 10.1021/acs.bioconjchem.3c00239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Inflammation-related diseases affect large populations of people in the world and cause substantial healthcare burdens, which results in significant costs in time, material, and labor. Preventing or relieving uncontrolled inflammation is critical for the treatment of these diseases. Herein, we report a new strategy for alleviating inflammation by macrophage reprogramming via targeted reactive oxygen species (ROS) scavenging and cyclooxygenase-2 (COX-2) downregulation. As a proof of concept, we synthesize a multifunctional compound named MCI containing a mannose-based macrophage targeting moiety, an indomethacin (IMC)-based segment for inhibiting COX-2, and a caffeic acid (CAF)-based section for ROS clearance. As revealed by a series of in vitro experiments, MCI could significantly attenuate the expression of COX-2 and the level of ROS, leading to M1 to M2 macrophage reprogramming, as evidenced by the reduction and the elevation in the levels of pro-inflammatory M1 markers and anti-inflammatory M2 markers, respectively. Furthermore, in vivo experiments show MCI's promising therapeutic effects on rheumatoid arthritis (RA). Our work illustrates the success of targeted macrophage reprogramming for inflammation alleviation, which sheds light on the development of new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Xiangjie Luo
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hui Xiong
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yuhang Jiang
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yifan Fan
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Cuicui Zuo
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Dongxia Chen
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Limin Chen
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hongyu Lin
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jinhao Gao
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
26
|
Tian H, Yan J, Zhang W, Li H, Jiang S, Qian H, Chen X, Dai X, Wang X. Cu-GA-coordination polymer nanozymes with triple enzymatic activity for wound disinfection and accelerated wound healing. Acta Biomater 2023:S1742-7061(23)00313-6. [PMID: 37270076 DOI: 10.1016/j.actbio.2023.05.048] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/21/2023] [Accepted: 05/29/2023] [Indexed: 06/05/2023]
Abstract
During the past few years, bacterial infection and oxidative stress have become important issues for wound healing. However, the emergence of numerous drug-resistant superbugs has had a serious impact on the treatment of infected wounds. Presently, the development of new nanomaterials has become one of the most important approaches to the treatment of drug-resistant bacterial infections. Herein, coordination polymer copper-gallic acid (Cu-GA) nanorods with multi-enzyme activity is successfully prepared for efficient wound treatment of bacterial infection, which can effectively promote wound healing. Cu-GA can be efficiently prepared by a simple solution method and had good physiological stability. Interestingly, Cu-GA shows enhanced multienzyme activity (peroxidase, glutathione peroxidase, and superoxide dismutase), which can produce a large number of reactive oxygen species (ROS) under acidic conditions while scavenging ROS under neutral conditions. In acidic environment, Cu-GA possesses POD (peroxidase)-like and glutathione peroxidase (GSH-Px)-like catalytic activities that is capable of killing bacteria; but in neutral environment, Cu-GA exhibits superoxide dismutase (SOD)-like catalytic activity that can scavenge ROS and promote wound healing. In vivo studies show that Cu-GA can promote wound infection healing and have good biosafety. Cu-GA contributes to the healing of infected wounds by inhibiting bacterial growth, scavenging reactive oxygen species, and promoting angiogenesis. STATEMENT OF SIGNIFICANCE: Cu-GA-coordinated polymer nanozymes with multienzyme activity were successfully prepared for efficient wound treatment of bacterial infection, which could effectively promote wound healing. Interestingly, Cu-GA exhibited enhanced multienzyme activity (peroxidase, glutathione peroxidase, and superoxide dismutase), which could produce a large number of reactive oxygen species (ROS) under acidic conditions and scavenge ROS under neutral conditions. In vitro and in vivo studies demonstrated that Cu-GA was capable of killing bacteria, controlling inflammation, and promoting angiogenesis.
Collapse
Affiliation(s)
- Haotian Tian
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, P.R. China; School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China
| | - Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, P. R. China
| | - Wei Zhang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China
| | - Huaixu Li
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, P.R. China
| | - Shouwei Jiang
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, P.R. China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China
| | - Xulin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Xingliang Dai
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, P.R. China.
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China; College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, P. R. China.
| |
Collapse
|
27
|
Zhang C, Li Q, Shan J, Xing J, Liu X, Ma Y, Qian H, Chen X, Wang X, Wu LM, Yu Y. Multifunctional two-dimensional Bi 2Se 3 nanodiscs for anti-inflammatory therapy of inflammatory bowel diseases. Acta Biomater 2023; 160:252-264. [PMID: 36805534 DOI: 10.1016/j.actbio.2023.02.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/18/2023]
Abstract
The overexpression of reactive oxygen and nitrogen species (RONS) in the colonic mucosa destroys the mucosa and its barrier, accelerating the occurrence of inflammatory bowel disease (IBD). The elimination of RONS from the inflammatory colon has proven effective in alleviating IBD. Although many nanoantioxidants have been developed, preparing robust and efficient nano-antioxidants remains challenging. Herein, by modifying bismuth selenide (Bi2Se3) nanodiscs with polyvinylpyrrolidone (PVP), a multifunctional nanozyme based on 2D nanomaterials was developed for the treatment of IBD. By eliminating multiple RONS, such as hydroxyl radicals (•OH), superoxide anions (O2-•), nitric oxide (NO), and Bi2Se3 nanodiscs enhanced cellular survival after H2O2 stimulation. As evidenced by colonic injury, reduced body weight, spleen index, and proinflammatory cytokine levels in mice, RONS clearance alleviated intestinal inflammation in a prevention and delay model of acute colitis. 16S rDNA amplicon sequencing reveals that Bi2Se3 nanodiscs had the potential to regulate intestinal flora, increase the proportion of Firmicutes to Bacteroidetes, inhibit Proteobacteria bacteria, and restore intestinal homeostasis. This study highlights the use of Bi2Se3 nanodiscs with excellent biocompatibility, multienzyme functionality, and RONS scavenging ability as treatments for IBD without apparent adverse effects. STATEMENT OF SIGNIFICANCE: RONS were efficiently scavenged by Bi2Se3 nanodiscs. Bi2Se3 nanodiscs could be as a promising and potentially safe theraeputic agent for IBD. The gut microbiota could be modulated by Bi2Se3 nanodiscs.
Collapse
Affiliation(s)
- Cong Zhang
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230026, China; School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Qingrong Li
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jie Shan
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Jianghao Xing
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Xiaoyan Liu
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yan Ma
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Xulin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China.
| | - Lian-Ming Wu
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Yue Yu
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230026, China.
| |
Collapse
|
28
|
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: 38] [Impact Index Per Article: 19.0] [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
|
29
|
Chang Y, Wu X, Lu S, Du J, Long Y, Zhu Y, Qin H. Engineered procyanidin-Fe nanoparticle alleviates intestinal inflammation through scavenging ROS and altering gut microbiome in colitis mice. Front Chem 2023; 11:1089775. [PMID: 37065822 PMCID: PMC10090317 DOI: 10.3389/fchem.2023.1089775] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an idiopathic chronic inflammatory bowel disease characterized by inflammation, intestinal barrier injury, and imbalance of gut microbiota. Excess accumulation of reactive oxygen species (ROS) is closely correlated with the development and reoccurrence of IBD. Previous researches demonstrate that procyanidin, as a natural antioxidant, exhibits strong ability of eliminating ROS, thus showing good therapeutic effects in the inflammation-related diseases. Non-etheless, its poor stability and solubility always limits the therapeutic outcomes. Here, we typically designed an antioxidant coordination polymer nanoparticle using the engineering of procyanidin (Pc) and free iron (Fe), named Pc-Fe nanozyme, for effectively scavenging ROS and further inhibiting inflammation while altering the gut microbiome for the treatment of colitis. Furthermore, in vitro experiments uncover that Pc-Fe nanoparticles exert strong multi biomimic activities, including peroxidase, and glutathione peroxidase, for the scavenging of ROS and protecting cells from oxidative injury. In addition, the colon accumulation of Pc-Fe nanozyme effectively protects the intestinal mucosa from oxidative damage while significantly downregulates pro-inflammatory factors, repairs the intestinal barriers and alternates gut microbiome after orally administrated in sodium dextran sulfate (DSS) induced colitis mice. The results collectively illustrate that the multienzyme mimicking Pc-Fe nanozyme owns high potential for treating IBD through scavenging ROS, inhibiting inflammation, repairing gut barriers and alternating gut microbiome, which further promising its clinical translation on IBD treatment and other ROS induced intestinal diseases.
Collapse
Affiliation(s)
- Yongliang Chang
- Shanghai Clinical College, Anhui Medical University, Shanghai, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
- Department of General Surgery, School of Medicine, Shanghai Tenth People’s Hospital Affiliated to Tongji University, Shanghai, China
| | - Xiawei Wu
- Shanghai Clinical College, Anhui Medical University, Shanghai, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
- Department of General Surgery, School of Medicine, Shanghai Tenth People’s Hospital Affiliated to Tongji University, Shanghai, China
| | - Shengwei Lu
- Shanghai Clinical College, Anhui Medical University, Shanghai, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
- Department of General Surgery, School of Medicine, Shanghai Tenth People’s Hospital Affiliated to Tongji University, Shanghai, China
| | - Jiahao Du
- Medical School of Nantong University, Nantong, China
| | - Yixiu Long
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Yixiu Long, ; Yefei Zhu, ; Huanlong Qin,
| | - Yefei Zhu
- Department of General Surgery, School of Medicine, Shanghai Tenth People’s Hospital Affiliated to Tongji University, Shanghai, China
- *Correspondence: Yixiu Long, ; Yefei Zhu, ; Huanlong Qin,
| | - Huanlong Qin
- Shanghai Clinical College, Anhui Medical University, Shanghai, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
- Department of General Surgery, School of Medicine, Shanghai Tenth People’s Hospital Affiliated to Tongji University, Shanghai, China
- Medical School of Nantong University, Nantong, China
- *Correspondence: Yixiu Long, ; Yefei Zhu, ; Huanlong Qin,
| |
Collapse
|
30
|
Du Y, Huo Y, Yang Q, Han Z, Hou L, Cui B, Fan K, Qiu Y, Chen Z, Huang W, Lu J, Cheng L, Cai W, Kang L. Ultrasmall iron-gallic acid coordination polymer nanodots with antioxidative neuroprotection for PET/MR imaging-guided ischemia stroke therapy. EXPLORATION (BEIJING, CHINA) 2023; 3:20220041. [PMID: 37323619 PMCID: PMC10190924 DOI: 10.1002/exp.20220041] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 12/21/2022] [Indexed: 06/17/2023]
Abstract
Oxidative stress from reactive oxygen species (ROS) is a reperfusion injury factor that can lead to cell damage and death. Here, ultrasmall iron-gallic acid coordination polymer nanodots (Fe-GA CPNs) were developed as antioxidative neuroprotectors for ischemia stroke therapy guided by PET/MR imaging. As proven by the electron spin resonance spectrum, the ultrasmall Fe-GA CPNs with ultrasmall size, scavenged ROS efficiently. In vitro experiments revealed that Fe-GA CPNs could protect cell viability after being treated with hydrogen peroxide (H2O2) and displayed the effective elimination of ROS by Fe-GA CPNs, which subsequently restores oxidation balance. When analyzing the middle cerebral artery occlusion model, the neurologic damage displayed by PET/MR imaging revealed a distinct recovery after treatment with Fe-GA CPNs, which was proved by 2,3,5-triphenyl tetrazolium chloride staining. Furthermore, immunohistochemistry staining indicated that Fe-GA CPNs inhibited apoptosis through protein kinase B (Akt) restoration, whereas western blot and immunofluorescence indicated the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) pathway following Fe-GA CPNs application. Therefore, Fe-GA CPNs exhibit an impressive antioxidative and neuroprotective role via redox homeostasis recovery by Akt and Nrf2/HO-1 pathway activation, revealing its potential for clinical ischemia stroke treatment.
Collapse
Affiliation(s)
- Yujing Du
- Department of Nuclear MedicinePeking University First HospitalBeijingChina
| | - Yan Huo
- Department of Nuclear MedicinePeking University First HospitalBeijingChina
| | - Qi Yang
- Department of Nuclear MedicinePeking University First HospitalBeijingChina
| | - Zhihui Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and TechnologySoochow UniversityJiangsuChina
| | - Linqian Hou
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and TechnologySoochow UniversityJiangsuChina
| | - Bixiao Cui
- Department of Radiology and Nuclear MedicineXuanwu Hospital Capital Medical UniversityBeijingChina
| | - Kevin Fan
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin‐MadisonWisconsinUSA
| | - Yongkang Qiu
- Department of Nuclear MedicinePeking University First HospitalBeijingChina
| | - Zhao Chen
- Department of Nuclear MedicinePeking University First HospitalBeijingChina
| | - Wenpeng Huang
- Department of Nuclear MedicinePeking University First HospitalBeijingChina
| | - Jie Lu
- Department of Radiology and Nuclear MedicineXuanwu Hospital Capital Medical UniversityBeijingChina
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and TechnologySoochow UniversityJiangsuChina
| | - Weibo Cai
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin‐MadisonWisconsinUSA
| | - Lei Kang
- Department of Nuclear MedicinePeking University First HospitalBeijingChina
| |
Collapse
|
31
|
Zhu L, Luo M, Zhang Y, Fang F, Li M, An F, Zhao D, Zhang J. Free radical as a double-edged sword in disease: Deriving strategic opportunities for nanotherapeutics. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
32
|
Zhang H, Sun X, Wang J, Dong M, Li L, Bai F, Xu K, Wang L. Sodium caseinate decorating on shellac nanoparticles as a stabilizer for the encapsulation of quercetin. Food Chem 2022; 395:133580. [DOI: 10.1016/j.foodchem.2022.133580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/04/2022]
|
33
|
Fang X, Hu JF, Hu QY, Li H, Sun ZJ, Xu Z, Zhang L. ROS-responsive resveratrol-loaded cyclodextrin nanomicelles reduce inflammatory osteolysis. Colloids Surf B Biointerfaces 2022; 219:112819. [PMID: 36137333 DOI: 10.1016/j.colsurfb.2022.112819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 10/31/2022]
Abstract
Bone loss in inflammatory disorders such as osteomyelitis, septic arthritis, and periodontitis is caused by excessive osteoclastic activity. Meanwhile, reactive oxygen species (ROS) have been identified as contributors to osteoclast differentiation, and the application of ROS scavengers has emerged as a promising strategy to protect against bone loss. Recently, resveratrol (RSV), a polyphenolic phytoalexin, has been demonstrated to inhibit osteoclastogenesis by scavenging ROS; however, the application of RSV as an antioxidant is limited by its low water solubility, structural instability, and short elimination half-life. In this study, we developed a PEGylated cyclodextrin (CD)-based nanoplatform (PCP) for local delivery of RSV as nanomicelles (RSV-NMs). In addition, polymer functionalization with phenylboronic acid ester in RSV-NMs successfully achieved ROS-responsive release of RSV. The RSV-NMs in a well-dispersed state possessed good biocompatibility as well as improved solubility and stability compared with RSV compound. In vitro, RSV-NMs significantly inhibited the formation of tartrate-resistant acid phosphatase (TRAP)-positive multinuclear cells and suppressed F-actin (filamentous actin) ring formation. Additionally, the mRNA expressions of osteoclastic marker genes, including matrix metalloprotein-9 (MMP-9), nuclear factor of activated T cells 1 (NFATc1), TRAP, and cathepsin K, were consequently downregulated in the presence of RSV-NMs. In vivo, RSV-NMs provided protection against LPS-induced bone destruction, as evidenced by a decreased number of osteoclasts, increased bone density, and reduced area of bone resorption. Taken together, these results indicate that our ROS-responsive RSV-NMs can be employed as a potential therapeutic agent for the treatment of inflammatory osteolysis.
Collapse
Affiliation(s)
- Xiaolin Fang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jun-Feng Hu
- School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, China
| | - Qing-Yun Hu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Han Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhigang Xu
- School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing 400715, China.
| | - Lu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
34
|
Guo L, Zhong S, Liu P, Guo M, Ding J, Zhou W. Radicals Scavenging MOFs Enabling Targeting Delivery of siRNA for Rheumatoid Arthritis Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202604. [PMID: 35661593 DOI: 10.1002/smll.202202604] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Indexed: 06/15/2023]
Abstract
Macrophages play essential roles in the progression of rheumatoid arthritis (RA), which are polarized into the pro-inflammatory M1 phenotype with significant oxidative stress and cytokines excretion. Herein, an active targeting nanomedicine based on metal-organic frameworks (MOFs) to re-educate the diseased macrophages for RA therapy is reported. The MOFs are prepared via coordination between tannic acid (TA) and Fe3+ , and anti-TNF-α siRNA is loaded via a simple sonication process, achieving high loading capacity comparable to cationic vectors. The MOFs show excellent biocompatibility, and enable rapid endo/lysosome escape of siRNA via the proton-sponge effect for effective cytokines down-regulation. Importantly, such nanomedicine displays intrinsic radicals scavenging capability to eliminate a broad spectrum of reactive oxygen and nitrogen species (RONS), which in turn repolarizes the M1 macrophages into anti-inflammatory M2 phenotypes for enhanced RA therapy in combination with siRNA. The MOFs are further modified with bovine serum albumin (BSA) to allow cascade RA joint and diseased macrophages targeted delivery. As a result, an excellent anti-RA efficacy is achieved in a collagen-induced arthritis mice model. This work provides a robust gene vector with great translational potential, and offers a vivid example of rationally designing MOF structure with multifunctionalities to synergize with its payload for enhanced disease treatment.
Collapse
Affiliation(s)
- Lina Guo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Shenghui Zhong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
- School of Medicine, Yichun University, Yichun, Jiangxi, 336000, China
| | - Peng Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Man Guo
- Academician Workstation, Changsha Medical University, Changsha, 410219, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
- Academician Workstation, Changsha Medical University, Changsha, 410219, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Changsha, Hunan, 410008, China
| |
Collapse
|
35
|
Yuan R, Li Y, Han S, Chen X, Chen J, He J, Gao H, Yang Y, Yang S, Yang Y. Fe-Curcumin Nanozyme-Mediated Reactive Oxygen Species Scavenging and Anti-Inflammation for Acute Lung Injury. ACS CENTRAL SCIENCE 2022; 8:10-21. [PMID: 35106369 PMCID: PMC8796308 DOI: 10.1021/acscentsci.1c00866] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Indexed: 05/16/2023]
Abstract
Pneumonia, such as acute lung injury (ALI), has been a type of lethal disease that is generally caused by uncontrolled inflammatory response and excessive generation of reactive oxygen species (ROS). Herein, we report Fe-curcumin-based nanoparticles (Fe-Cur NPs) with nanozyme functionalities in guiding the intracellular ROS scavenging and meanwhile exhibiting anti-inflammation efficacy for curing ALI. The nanoparticles are noncytotoxic when directing these biological activities. Mechanism studies for the anti-inflammation aspects of Fe-Cur NPs were systematically carried out, in which the infected cells and tissues were alleviated through downregulating levels of several important inflammatory cytokines (such as TNF-α, IL-1β, and IL-6), decreasing the intracellular Ca2+ release, inhibiting NLRP3 inflammasomes, and suppressing NF-κB signaling pathways. In addition, we performed both the intratracheal and intravenous injection of Fe-Cur NPs in mice experiencing ALI and, importantly, found that the accumulation of such nanozymes was enhanced in lung tissue (better than free curcumin drugs), demonstrating its promising therapeutic efficiency in two different administration methods. We showed that the inflammation reduction of Fe-Cur NPs was effective in animal experiments and that ROS scavenging was also effectively achieved in lung tissue. Finally, we revealed that Fe-Cur NPs can decrease the level of macrophage cells (CD11bloF4/80hi) and CD3+CD45+ T cells in mice, which could help suppress the inflammation cytokine storm caused by ALI. Overall, this work has developed the strategy of using Fe-Cur NPs as nanozymes to scavenge intracellular ROS and as an anti-inflammation nanodrugs to synergistically cure ALI, which may serve as a promising therapeutic agent in the clinical treatment of this deadly disease. Fe-Cur NP nanozymes were designed to attenuate ALI by clearing intracellular ROS and alleviating inflammation synergistically. Relevant cytokines, inflammasomes, and signaling pathways were studied.
Collapse
Affiliation(s)
- Renyikun Yuan
- College
of Pharmacy, Guangxi University of Chinese
Medicine, Nanning 530000, China
- College
of Pharmacy, Jiangxi University of Traditional
Chinese Medicine, Nanchang 330004, China
| | - Yuqing Li
- Department
of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Shan Han
- College
of Pharmacy, Guangxi University of Chinese
Medicine, Nanning 530000, China
| | - Xinxin Chen
- College
of Pharmacy, Guangxi University of Chinese
Medicine, Nanning 530000, China
| | - Jingqi Chen
- Institute
of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jia He
- College
of Pharmacy, Guangxi University of Chinese
Medicine, Nanning 530000, China
| | - Hongwei Gao
- College
of Pharmacy, Guangxi University of Chinese
Medicine, Nanning 530000, China
| | - Yang Yang
- Department
of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- School
of Materials Science and Engineering, Tongji
University, Shanghai 201804, China
| | - Shilin Yang
- College
of Pharmacy, Guangxi University of Chinese
Medicine, Nanning 530000, China
| | - Yu Yang
- Institute
of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
36
|
Zheng N, Fu Y, Liu X, Zhang Z, Wang J, Mei Q, Wang X, Deng G, Lu J, Hu J. Tumor microenvironment responsive self-cascade catalysis for synergistic chemo/chemodynamic therapy by multifunctional biomimetic nanozymes. J Mater Chem B 2022; 10:637-645. [PMID: 34991154 DOI: 10.1039/d1tb01891d] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chemodynamic therapy (CDT) is an emerging approach to treat cancer based on the tumor microenvironment (TME), but its limited content of endogenous hydrogen peroxide (H2O2) weakens the anticancer effects. Herein, a multifunctional biomimetic nanozyme (Se@SiO2-Mn@Au/DOX, named as SSMA/DOX) is fabricated, which undergoes TME responsive self-cascade catalysis to facilitate MRI guided enhanced chemo/chemodynamic therapy. The SSMA/DOX nanocomposites (NCs) responsively degrade in acidic conditions of tumor to release Se, DOX, Au and Mn2+. Mn2+ not only enables MRI to guided therapy, but also catalyzes the endogenous H2O2 into hydroxyl radical (˙OH) for CDT. In addition, the Au NPs continuously catalyze glucose to generate H2O2, enhancing CDT by supplementing a sufficiently reactive material and cutting off the energy supply of the tumor by consuming glucose. Simultaneously, Se enhances the chemotherapy of doxorubicin hydrochloride (DOX) and CDT by upregulating ROS in the tumor cells, achieving remarkable inhibition effect towards tumor. Moreover, SSMA/DOX NCs have good biocompatibility and degradability, which avoid long-term toxicity and side effects. Overall, the degradable SSMA/DOX NCs provide an innovative strategy for tumor microenvironment responsive self-cascade catalysis to enhance tumor therapy.
Collapse
Affiliation(s)
- Nannan Zheng
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China. .,College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China.
| | - Yang Fu
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai 201600, China.,Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201600, China
| | - Xijian Liu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Ziwen Zhang
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Jinxia Wang
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Qixiang Mei
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai 201600, China.,Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201600, China
| | - Xingyan Wang
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Guoying Deng
- Trauma Center, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201620, China
| | - Jie Lu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Junqing Hu
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China. .,Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
37
|
Zhang K, Wang X, Tian M, Gou Z, Zuo Y. The diversity of the coordination bond generated a POSS-based fluorescent probe for the reversible detection of Cu(II), Fe(III) and amino acids. J Mater Chem B 2021; 9:9744-9753. [PMID: 34787631 DOI: 10.1039/d1tb01947c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In recent years, it has been found that Cu2+, Fe3+, and amino acids play an irreplaceable and subtle role in organisms and have attracted the considerable attention of many researchers. Therefore, it is vital to design visual indicators to reveal the relationships between metal ions and amino acids. However, there have been few reports on this vigorous subject. Fortunately, based on the different coordination effects between metal ions and boron groups, we have designed an accessible fluorescent probe (PSI-A). Borane was introduced as an ion-sensitive group to form a novel POSS-based fluorescent probe, which achieves fascinating performance, in situ dynamic multiple detection, excellent photostability, and enervative biological toxicity. PSI-A exhibited predominant selectivity and sensitivity to Cu2+/amino acids and Fe3+/amino acids sequence reactions in HepG2 cells and zebrafish. The fluorescence of PSI-A was quenched by Cu2+, which can be recovered by adding Asp, Ser, Arg, Ace or Trp. Additionally, the fluorescence of PSI-A quenched by Fe3+ can be restored after adding Asp. PSI-A is available to monitor Cu2+/amino acids and Fe3+/amino acids sequence reactions and can be repeated for at least three consecutive cycles without a fatigued performance. Therefore, this multifunctional fluorescent probe may have prospective application potentials in the biological field.
Collapse
Affiliation(s)
- Kun Zhang
- School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Shandong 250022, P. R. China.
| | - Xiaoni Wang
- School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Shandong 250022, P. R. China.
| | - Minggang Tian
- School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Shandong 250022, P. R. China.
| | - Zhiming Gou
- School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Shandong 250022, P. R. China.
| | - Yujing Zuo
- School of Chemistry and Chemical Engineering, School of Materials Science and Engineering, University of Jinan, Shandong 250022, P. R. China.
| |
Collapse
|
38
|
Yang P, Zhang J, Xiang S, Jin Z, Zhu F, Wang T, Duan G, Liu X, Gu Z, Li Y. Green Nanoparticle Scavengers against Oxidative Stress. ACS APPLIED MATERIALS & INTERFACES 2021; 13:39126-39134. [PMID: 34383476 DOI: 10.1021/acsami.1c12176] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The usage of exogenous antioxidant materials to relieve oxidative stress offers an important strategy for the therapy of oxidative stress-induced injuries. However, the fabrication processes toward the antioxidant materials usually require the involvement of extra metal ions and organic agents, as well as sophisticated purification steps, which might cause tremendous environmental stress and induce unpredictable side effects in vivo. To address these issues, herein, we proposed a novel strategy to fabricate green nanoparticles for efficiently modulating oxidative stress, which was facilely prepared from tea polyphenol extracts (originated from green tea) via a green enzymatic polymerization-based chemistry method. The resulting nanoparticles possessed a uniform spherical morphology and good stability in water and biomedium and demonstrated excellent radical scavenging properties. These nanoparticle scavengers could effectively prevent intracellular oxidative damage, accelerate wound recovery, and protect the kidneys from reactive oxygen species damaging in the acute kidney injury model. We hope this work will inspire the further development of more types of green nanoparticles for antioxidant therapies via similar synthetic strategies using green biomass materials.
Collapse
Affiliation(s)
- Peng Yang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Jianhua Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Siying Xiang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zhekai Jin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Fang Zhu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Tianyou Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Gaigai Duan
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Materials Science and Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Xianhu Liu
- National Engineering Research Center for Advanced Polymer Processing Technology, Zhengzhou University, Zhengzhou 450002, China
| | - Zhipeng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Yiwen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
39
|
Chen Q, Ding F, Zhang S, Li Q, Liu X, Song H, Zuo X, Fan C, Mou S, Ge Z. Sequential Therapy of Acute Kidney Injury with a DNA Nanodevice. NANO LETTERS 2021; 21:4394-4402. [PMID: 33998787 DOI: 10.1021/acs.nanolett.1c01044] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The high demand for acute kidney injury (AKI) therapy calls the development of multifunctional nanomedicine for renal management with programmable pharmacokinetics. Here, we developed a renal-accumulating DNA nanodevice with exclusive kidney retention for longitudinal protection of AKI in different stages in a renal ischemia-reperfusion (I/R) model. Due to the prolonged kidney retention time (>12 h), the ROS-sensitive nucleic acids of the nanodevice could effectively alleviate oxidative stress by scavenging ROS in stage I, and then the anticomplement component 5a (aC5a) aptamer loaded nanodevice could sequentially suppress the inflammatory responses by blocking C5a in stage II, which is directly related to the cytokine storm. This sequential therapy provides durable and pathogenic treatment of kidney dysfunction based on successive pathophysiological events induced by I/R, which holds great promise for renal management and the suppression of the cytokine storm in more broad settings including COVID-19.
Collapse
Affiliation(s)
- Qian Chen
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Fei Ding
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuangye Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaoguo Liu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haiyun Song
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine and School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shan Mou
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhilei Ge
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|