1
|
Geng S, Fang B, Wang K, Wang F, Zhou Y, Hou Y, Iqbal MZ, Chen Y, Yu Z. Polydopamine Nanoformulations Induced ICD and M1 Phenotype Macrophage Polarization for Enhanced TNBC Synergistic Photothermal Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59814-59832. [PMID: 39450881 DOI: 10.1021/acsami.4c11594] [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: 10/26/2024]
Abstract
Photothermal therapy (PTT) is a promising technology that can achieve the thermal ablation of tumors and induce immunogenic cell death (ICD). However, relying solely on the antitumor immune responses caused by PTT-induced ICD is insufficient to suppress tumor metastasis and recurrence effectively. Fortunately, multifunctional nanoformulation-based synergistic photothermal immunotherapy can eliminate primary and metastatic tumors and inhibit tumor recurrence for a long time. Herein, we select polydopamine (PDA) nanoparticles to serve as the carrier for our nanomedicine as well as a potent photothermal agent and modulator of macrophage polarization. PDA nanoparticles are loaded with the insoluble immune adjuvant Imiquimod (R837) to construct PDA(R837) nanoformulations. These straightforward yet highly effective nanoformulations demonstrate excellent performance, allowing for successful triple-negative breast cancer (TNBC) treatment through synergistic photothermal immunotherapy. Moreover, experimental results showed that PDA(R837) implementation of PTT is effective in the thermal ablation of primary tumors while causing ICD and releasing R837, further promoting dendritic cell (DC) maturation and activating the systemic antitumor immune response. Furthermore, PDA(R837) nanoformulations inhibit tumor metastasis and recurrence and achieve M1 phenotype macrophage polarization, achieving long-term and excellent antitumor efficacy. Therefore, the structurally simple PDA(R837) nanoformulations provide cancer treatment and have excellent clinical translational application prospects.
Collapse
Affiliation(s)
- Siqi Geng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Baoru Fang
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Ke Wang
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Fang Wang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Yiqing Zhou
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Yike Hou
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P. R. China
| | - M Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P. R. China
| | - Yanping Chen
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Zhangsen Yu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| |
Collapse
|
2
|
Liu H, Zhuo R, Zou C, Xu S, Cai X, Ge Y, Liu G, Wu C, Dai C, Li J, Fan Z, Yang L, Li Y. RVG-peptide-camouflaged iron-coordinated engineered polydopamine nanoenzyme with ROS scavenging and inhibiting inflammatory response for ischemic stroke therapy. Int J Biol Macromol 2024; 282:136778. [PMID: 39442842 DOI: 10.1016/j.ijbiomac.2024.136778] [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: 06/11/2024] [Revised: 09/30/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Stroke is one of the most common causes of death and disability. In addition, most neuroprotective agents fail to rescue neurons from cerebral ischemic insults due to their poor ability to penetrate the blood-brain barrier (BBB). Here, the tailored engineered nanoenzyme has been successfully synthesized by coordination-driven co-assembly of dopamine (DA) and iron ion (Fe3+), which is subsequently camouflaged by neuron-specific rabies viral glycoprotein (RVG) peptide to scavenge reactive oxygen species (ROS) and inhibit inflammatory response in damaged neuron for the efficient therapy of ischemic stroke. The resulting nanoenzyme with good biocompatibility, core-shell structure, and suitable diameter can nondestructively cross the BBB and then internalize into the damaged neuron through the camouflaging and homologous targeted strategy of neuron-specific RVG peptide. After intravenous injection into transient middle cerebral artery occlusion (tMCAO) mouse models, nanoenzyme exerted a significant neuroprotective effect, resulting in a 50 % reduction in neurological scores and an approximate 33 % decrease in cerebral infarction volume. Interestingly, such nanoenzyme can eliminate free radicals, reduce neuroinflammation, enhance BBB integrity, improve mitochondrial function, and inhibit neuronal ferroptosis. Taken together, this well-designed nanoenzyme with its excellent biocompatibility and well-understood mechanisms holds promise a robust therapy for ischemic stroke.
Collapse
Affiliation(s)
- Heng Liu
- Department of Pharmacy, Xiamen Medical College & The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China; Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Rengong Zhuo
- Department of Pharmacy, Xiamen Medical College & The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Chuanyang Zou
- Department of Pharmacy, Xiamen Medical College & The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Shuyu Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xinying Cai
- Department of Pharmacy, Xiamen Medical College & The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yuxue Ge
- Department of Pharmacy, Xiamen Medical College & The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Chuang Wu
- Department of Pharmacy, Xiamen Medical College & The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China.
| | - Cuilian Dai
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen 361002, China.
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China.
| | - Zhongxiong Fan
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China; School of Pharmaceutical Sciences, Institute of Materia Medica, Xinjiang University, Urumqi 830017, China.
| | - Lichao Yang
- Department of Pharmacy, Xiamen Medical College & The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China.
| | - Ying Li
- Department of Pharmacy, Xiamen Medical College & The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
3
|
Zeng F, Qin Y, Nijiati S, Liu Y, Ye J, Shen H, Cai J, Xiong H, Shi C, Tang L, Yu C, Zhou Z. Ultrasmall Nanodots with Dual Anti-Ferropototic Effect for Acute Kidney Injury Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403305. [PMID: 39159052 PMCID: PMC11497046 DOI: 10.1002/advs.202403305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/28/2024] [Indexed: 08/21/2024]
Abstract
Ferroptosis is known to mediate the pathogenesis of chemotherapeutic drug-induced acute kidney injury (AKI); however, leveraging the benefits of ferroptosis-based treatments for nephroprotection remains challenging. Here, ultrasmall nanodots, denoted as FerroD, comprising the amphiphilic conjugate (tetraphenylethylene-L-serine-deferoxamine, TPE-lys-Ser-DFO (TSD)) and entrapped ferrostatin-1 are designed. After being internalized through kidney injury molecule-1-mediated endocytosis, FerroD can simultaneously remove the overloaded iron ions and eliminate the overproduction of lipid peroxides by the coordination-disassembly mechanisms, which collectively confer prominent inhibition efficiency of ferroptosis. In cisplatin (CDDP)-induced AKI mice, FerroD equipped with dual anti-ferroptotic ability can provide long-term nephroprotective effects. This study may shed new light on the design and clinical translation of therapeutics targeting ferroptosis for various ferroptosis-related kidney diseases.
Collapse
Affiliation(s)
- Fantian Zeng
- State Key Laboratory of Vaccines for Infectious DiseasesXiang An Biomedicine LaboratorySchool of Public HealthShenzhen Research Institute of Xiamen UniversityXiamen UniversityXiamen361102China
| | - Yatong Qin
- State Key Laboratory of Vaccines for Infectious DiseasesXiang An Biomedicine LaboratorySchool of Public HealthShenzhen Research Institute of Xiamen UniversityXiamen UniversityXiamen361102China
| | - Sureya Nijiati
- State Key Laboratory of Vaccines for Infectious DiseasesXiang An Biomedicine LaboratorySchool of Public HealthShenzhen Research Institute of Xiamen UniversityXiamen UniversityXiamen361102China
| | - Yangtengyu Liu
- Department of Rheumatology and ImmunologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Jinmin Ye
- State Key Laboratory of Vaccines for Infectious DiseasesXiang An Biomedicine LaboratorySchool of Public HealthShenzhen Research Institute of Xiamen UniversityXiamen UniversityXiamen361102China
| | - Huaxiang Shen
- State Key Laboratory of Vaccines for Infectious DiseasesXiang An Biomedicine LaboratorySchool of Public HealthShenzhen Research Institute of Xiamen UniversityXiamen UniversityXiamen361102China
| | - Jiayuan Cai
- State Key Laboratory of Vaccines for Infectious DiseasesXiang An Biomedicine LaboratorySchool of Public HealthShenzhen Research Institute of Xiamen UniversityXiamen UniversityXiamen361102China
| | - Hehe Xiong
- State Key Laboratory of Vaccines for Infectious DiseasesXiang An Biomedicine LaboratorySchool of Public HealthShenzhen Research Institute of Xiamen UniversityXiamen UniversityXiamen361102China
| | - Changrong Shi
- State Key Laboratory of Vaccines for Infectious DiseasesXiang An Biomedicine LaboratorySchool of Public HealthShenzhen Research Institute of Xiamen UniversityXiamen UniversityXiamen361102China
- Departments of Diagnostic Radiology, SurgeryChemical and Biomolecular Engineeringand Biomedical EngineeringYong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingapore119074Singapore
| | | | - Chunyang Yu
- School of Chemistry and Chemical EngineeringState Key Laboratory of Metal Matrix CompositesShanghai Jiao Tong University800 Dongchuan RoadShanghai200240China
| | - Zijian Zhou
- State Key Laboratory of Vaccines for Infectious DiseasesXiang An Biomedicine LaboratorySchool of Public HealthShenzhen Research Institute of Xiamen UniversityXiamen UniversityXiamen361102China
| |
Collapse
|
4
|
Kwon YS, Han Z. Advanced nanomedicines for the treatment of age-related macular degeneration. NANOSCALE 2024; 16:16769-16790. [PMID: 39177654 DOI: 10.1039/d4nr01917b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
The critical and unmet medical need for novel therapeutic advancements in the treatment of age-related macular degeneration (AMD) cannot be overstated, particularly given the aging global population and the increasing prevalence of this condition. Current AMD therapy involves intravitreal treatments that require monthly or bimonthly injections to maintain optimal efficacy. This underscores the necessity for improved approaches, prompting recent research into developing advanced drug delivery systems to prolong the intervals between treatments. Nanoparticle-based therapeutic approaches have enabled the controlled release of drugs, targeted delivery of therapeutic materials, and development of smart solutions for the harsh microenvironment of diseased tissues, offering a new perspective on ocular disease treatment. This review emphasizes the latest pre-clinical treatment options in ocular drug delivery to the retina and explores the advantages of nanoparticle-based therapeutic approaches, with a focus on AMD, the leading cause of irreversible blindness in the elderly.
Collapse
Affiliation(s)
- Yong-Su Kwon
- Department of Ophthalmology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | - Zongchao Han
- Department of Ophthalmology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
5
|
Eom T, Ozlu B, Ivanová L, Lee S, Lee H, Krajčovič J, Shim BS. Multifunctional Natural and Synthetic Melanin for Bioelectronic Applications: A Review. Biomacromolecules 2024; 25:5489-5511. [PMID: 39194016 DOI: 10.1021/acs.biomac.4c00494] [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: 08/29/2024]
Abstract
Emerging material interest in bioelectronic applications has highlighted natural melanin and its derivatives as promising alternatives to conventional synthetic conductors. These materials, traditionally noted for their adhesive, antioxidant, biocompatible, and biodegradable properties, have barely been used as conductors due to their extremely low electrical activities. However, recent studies have demonstrated good conductive properties in melanin materials that promote electronic-ionic hybrid charge transfer, attributed to the formation of an extended conjugated backbone. This review examines the multifunctional properties of melanin materials, focusing on their chemical and electrochemical synthesis and their resulting structure-property-function relationship. The wide range of bioelectronic applications will also be presented to highlight their importance and potential to expand into new design concepts for high-performance electronic functional materials. The review concludes by addressing the current challenges in utilizing melanin for biodegradable bioelectronics, providing a perspective on future developments.
Collapse
Affiliation(s)
- Taesik Eom
- Program in Biomedical Science & Engineering, Inha University, 100 Inharo, Michuhol-gu, Incheon 22212, South Korea
- Department of Chemical Engineering, Inha University, 100 Inharo, Michuhol-gu, Incheon 22212, South Korea
- KIURI Center for Hydrogen Based Next Generation Mechanical System, Inha University, 36 Gaetbeol-ro, Yeonsu-gu, Incheon 21999, South Korea
| | - Busra Ozlu
- Program in Biomedical Science & Engineering, Inha University, 100 Inharo, Michuhol-gu, Incheon 22212, South Korea
- Department of Chemical Engineering, Inha University, 100 Inharo, Michuhol-gu, Incheon 22212, South Korea
| | - Lucia Ivanová
- Faculty of Chemistry, Brno University of Technology, Purkyňova 118, CZ-612 00 Brno, Czech Republic
| | - Seunghyeon Lee
- Program in Biomedical Science & Engineering, Inha University, 100 Inharo, Michuhol-gu, Incheon 22212, South Korea
- Department of Chemical Engineering, Inha University, 100 Inharo, Michuhol-gu, Incheon 22212, South Korea
| | - HyeonJeong Lee
- Program in Biomedical Science & Engineering, Inha University, 100 Inharo, Michuhol-gu, Incheon 22212, South Korea
| | - Jozef Krajčovič
- Faculty of Chemistry, Brno University of Technology, Purkyňova 118, CZ-612 00 Brno, Czech Republic
| | - Bong Sup Shim
- Program in Biomedical Science & Engineering, Inha University, 100 Inharo, Michuhol-gu, Incheon 22212, South Korea
- Department of Chemical Engineering, Inha University, 100 Inharo, Michuhol-gu, Incheon 22212, South Korea
| |
Collapse
|
6
|
Minibayeva FV, Rassabina AE, Zakirjanova GF, Fedorov NS, Khabibrakhmanova VR, Galeeva EI, Kuznetsova EA, Malomouzh AI, Petrov AM. Protective properties of melanin from lichen Lobaria pulmonaria (L.) HOFFM. In models of oxidative stress in skeletal muscle. Fitoterapia 2024; 177:106127. [PMID: 39019238 DOI: 10.1016/j.fitote.2024.106127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/16/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
Melanin is a dark pigment from the group of phenolic or indole polymers with inherent biocompatibility and antioxidant capacity. In extremophilic lichen Lobaria pulmonaria, melanin is responsible for protective properties against hostile environments. Herein, the ability of melanin extracted from L. pulmonaria to counteract oxidative stress and related damages was studied in the mouse diaphragm, the main respiratory muscle. Initial in vitro experiments demonstrated ultraviolet (UV)-absorbing, antioxidant and metal chelating activities of melanin. This melanin can form nanoparticles and stabile colloidal system at concentration of 5 μg/ml. Pretreatment of the muscle with melanin (5 μg/ml) markedly reduced UV-induced increase in intracellular and extracellular reactive oxygen species (ROS) as well as antimycin A-mediated enhancement in mitochondrial ROS production accompanied by lipid peroxidation and membrane asymmetry loss. In addition, melanin attenuated suppression of neuromuscular transmission and alterations of contractile responses provoked by hydrogen peroxide. Thus, this study shed the light on the perspectives of the application of a lichen melanin as a protective component for treatment of skeletal muscle disorders, which are accompanied with an increased ROS production.
Collapse
Affiliation(s)
- Farida V Minibayeva
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia.
| | - Anna E Rassabina
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia
| | - Guzalia F Zakirjanova
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia
| | - Nikita S Fedorov
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia
| | - Venera R Khabibrakhmanova
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia
| | - Ekaterina I Galeeva
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia
| | - Eva A Kuznetsova
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia
| | - Artem I Malomouzh
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia; Kazan National Research Technical University, 10, K. Marx St., Kazan 420111, Russia
| | - Alexey M Petrov
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, Box 30, Kazan 420111, Russia; Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia; Kazan State Mediсal University, 49 Butlerova Street, Kazan 420012, Russia.
| |
Collapse
|
7
|
Xin W, Gong S, Chen Y, Yao M, Qin S, Chen J, Zhang A, Yu W, Zhou S, Zhang B, Gu J, Zhao J, Huang Y. Self-Assembling P38 Peptide Inhibitor Nanoparticles Ameliorate the Transition from Acute to Chronic Kidney Disease by Suppressing Ferroptosis. Adv Healthc Mater 2024; 13:e2400441. [PMID: 38775779 DOI: 10.1002/adhm.202400441] [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: 02/04/2024] [Revised: 05/13/2024] [Indexed: 05/28/2024]
Abstract
Accumulating evidence highlights p38 as a crucial factor highly activated during the process of acute kidney injury (AKI), but the application of p38 inhibitor in AKI is quite limited due to the low efficiency and poor kidney-targeting ability. Herein, a novel self-assembling peptide nanoparticle with specific p38-inhibiting activity is constructed, which linked mitogen-activated protein kinase kinase 3b (MKK3b), the functional domain of p38, with the cell-penetrating TAT sequence, ultimately self-assembling into TAT-MKK3b nanoparticles (TMNPs) through tyrosinase oxidation. Subsequent in vitro and in vivo studies demonstrated that TMNPs preferably accumulated in the renal tubular epithelial cells (RTECs) through forming protein coronas by binding to albumin, and strongly improved the reduced renal function of ischemia-reperfusion injury (IRI)-induced AKI and its transition to chronic kidney disease (CKD). Mechanically, TMNPs inhibited ferroptosis via its solute carrier family 7 member 11 (SLC7A11)/glutathione peroxidase 4 (GPX4) axis-inducing capacity and synergistic potent antioxidant property in AKI. The findings indicated that the multifunctional TMNPs exhibited renal targeting, ROS-scavenging, and ferroptosis-mitigating capabilities, which may serve as a promising therapeutic agent for the treatment of AKI and its progression to CKD.
Collapse
Affiliation(s)
- Wang Xin
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Shuiqin Gong
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Yin Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Mengying Yao
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Shaozong Qin
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jing Chen
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Aihong Zhang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Wenrui Yu
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Siyan Zhou
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Bo Zhang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jun Gu
- State Key Laboratory of Protein and Plant Gene Research, College of Life Science, Peking University, Beijing, 100871, China
| | - Jinghong Zhao
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Yinghui Huang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| |
Collapse
|
8
|
Liu R, Zhou T, Li X, Zou Q, Yu J, Ye J, Wang W, Zhou Y, Sun SK. A Non-Metallic Nanozyme Ameliorates Pulmonary Hypertension Through Inhibiting ROS/TGF-β1 Signaling. Adv Healthc Mater 2024:e2401909. [PMID: 39155419 DOI: 10.1002/adhm.202401909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/01/2024] [Indexed: 08/20/2024]
Abstract
Pulmonary hypertension (PH) is a life-threatening cardiovascular disease with a lack of effective treatment options. Nanozymes, though promising for PH therapy, pose safety risks due to their metallic nature. Here, a non-metallic nanozyme is reported for the treatment of monocrotaline (MCT)-induced PH with a therapeutic mechanism involving the ROS/TGF-β1 signaling. The synthesized melanin-polyvinylpyrrolidone-polyethylene glycol (MPP) nanoparticles showcase ultra-small size, excellent water solubility, high biocompatibility, and remarkable antioxidant capacity. The MPP nanoparticles are capable of effectively eliminating ROS in isolated pulmonary artery smooth muscle cells (PASMCs) from PH rats, and significantly reduce PASMC proliferation and migration. In vivo results from a PH model demonstrate that MPP nanoparticles significantly increase pulmonary artery acceleration time, decrease wall thickening and PCNA expression in lung tissues, as evidenced by echocardiograpy, histology and immunoblot analysis. Additionally, MPP nanoparticles treatment improve running capacity, decrease Fulton index, and attenuate right ventricular fibrosis in MCT-PH rats by using treadmill test, picrosirius red, and trichrome Masson staining. Further transcriptomic and biochemical analyses reveal that inhibiting ROS-driven activation of TGF-β1 in the PA is the mechanism by which MPP nanoparticles exert their therapeutic effect. This study provides a novel approach for treating PH with non-metallic nanozymes based on a well-understood mechanism.
Collapse
Affiliation(s)
- Ruxia Liu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Ting Zhou
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
- Department of CT, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, 710068, China
| | - Xinsheng Li
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| | - Quan Zou
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, 300203, China
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jiaojiao Yu
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| | - Jingjing Ye
- Trauma Treatment Center, Peking University People's Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine, Beijing, 100044, China
| | - Wenhui Wang
- Department of Endocrinology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yan Zhou
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| |
Collapse
|
9
|
Liu D, Sun S, Qiao H, Xin Q, Zhou S, Li L, Song N, Zhang L, Chen Q, Tian F, Mu X, Zhang S, Zhang J, Guo M, Wang H, Zhang XD, Zhang R. Ce 12V 6 Clusters with Multi-Enzymatic Activities for Sepsis Treatment. Adv Healthc Mater 2024:e2401581. [PMID: 39129228 DOI: 10.1002/adhm.202401581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/22/2024] [Indexed: 08/13/2024]
Abstract
Artificial enzymes, especially nanozymes, have attracted wide attention due to their controlled catalytic activity, selectivity, and stability. The rising Cerium-based nanozymes exhibit unique SOD-like activity, and Vanadium-based nanozymes always hold excellent GPx-like activity. However, most inflammatory diseases involve polymerase biocatalytic processes that require multi-enzyme activities. The nanocomposite can fulfill multi-enzymatic activity simultaneously, but large nanoparticles (>10 nm) cannot be excreted rapidly, leading to biosafety challenges. Herein, atomically precise Ce12V6 clusters with a size of 2.19 nm are constructed. The Ce12V6 clusters show excellent glutathione peroxidase (GPx) -like activity with a significantly lower Michaelis-Menten constant (Km, 0.0125 mM versus 0.03 mM of natural counterpart) and good activities mimic superoxide dismutase (SOD) and peroxidase (POD). The Ce12V6 clusters exhibit the ability to scavenge the ROS including O2 ·- and H2O2 via the cascade reactions of multi-enzymatic activities. Further, the Ce12V6 clusters modulate the proinflammatory cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) and consequently rescue the multi-organ failure in the lipopolysaccharide (LPS)-induced sepsis mouse model. With excellent biocompatibility, the Ce12V6 clusters show promise in the treatment of sepsis.
Collapse
Affiliation(s)
- Di Liu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Si Sun
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Huanhuan Qiao
- 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
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Sufei Zhou
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Lingxia Li
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Nan Song
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin, 300384, 18, China
| | - Lijie Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Qi Chen
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Fangzhen Tian
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiaoyu Mu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Shaofang Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jing Zhang
- Department of Cardiology Tianjin Chest Hospital, Tianjin University, Tianjin, 300222, China
| | - Meili Guo
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin, 300384, 18, 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
| | - Ruiping Zhang
- The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
10
|
Wang T, Zhang J, Chen Z, Zhang R, Duan G, Wang Z, Chen X, Gu Z, Li Y. Sonochemical Synthesis of Natural Polyphenolic Nanoparticles for Modulating Oxidative Stress. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401731. [PMID: 38682736 DOI: 10.1002/smll.202401731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/01/2024] [Indexed: 05/01/2024]
Abstract
Natural polyphenolic compounds play a vital role in nature and are widely utilized as building blocks in the fabrication of emerging functional nanomaterials. Although diverse fabrication methodologies are developed in recent years, the challenges of purification, uncontrollable reaction processes and additional additives persist. Herein, a modular and facile methodology is reported toward the fabrication of natural polyphenolic nanoparticles. By utilizing low frequency ultrasound (40 kHz), the assembly of various natural polyphenolic building blocks is successfully induced, allowing for precise control over the particle formation process. The resulting natural polyphenolic nanoparticles possessed excellent in vitro antioxidative abilities and in vivo therapeutic effects in typical oxidative stress models including wound healing and acute kidney injury. This study opens new avenues for the fabrication of functional materials from naturally occurring building blocks, offering promising prospects for future advancements in this field.
Collapse
Affiliation(s)
- Tianyou Wang
- 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
| | - Zhan Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Rong Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Gaigai Duan
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Materials Science and Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Zhao Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Xianchun Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, 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
|
11
|
Terranova ML. Physiological Roles of Eumelanin- and Melanogenesis-Associated Diseases: A Look at the Potentialities of Engineered and Microbial Eumelanin in Clinical Practice. Bioengineering (Basel) 2024; 11:756. [PMID: 39199714 PMCID: PMC11351163 DOI: 10.3390/bioengineering11080756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
This paper aims to highlight the physiological actions exerted by eumelanin present in several organs/tissues of the human body and to rationalise the often conflicting functional roles played by this biopolymer on the basis of its peculiar properties. Besides pigmentary disorders, a growing number of organ injuries and degenerative pathologies are presently ascribed to the modification of physiological eumelanin levels in terms of alterations in its chemical/structural features, and of a partial loss or uneven distribution of the pigment. The present review analyses the more recent research dedicated to the physiological and pathological actions of eumelanin and provides an insight into some melanogenesis-associated diseases of the skin, eye, ear, and brain, including the most significant neurodegenerative disorders. Also described are the potentialities of therapies based on the localised supply of exogeneous EU and the opportunities that EU produced via synthetic biology offers in order to redesign therapeutical and diagnostic applications.
Collapse
Affiliation(s)
- Maria Letizia Terranova
- Dip.to di Scienze e Tecnologie Chimiche, Università degli Studi di Roma "Tor Vergata", 00133 Roma, Italy
| |
Collapse
|
12
|
Zhang W, Chan C, Zhang K, Qin H, Yu BY, Xue Z, Zheng X, Tian J. Discovering a New Drug Against Acute Kidney Injury by Using a Tailored Photoacoustic Imaging Probe. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311397. [PMID: 38221651 DOI: 10.1002/adma.202311397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/29/2023] [Indexed: 01/16/2024]
Abstract
Acute kidney injury (AKI) has become an increasing concern for patients due to the widespread clinical use of nephrotoxic drugs. Currently, the early diagnosis of AKI is still challenging and the available therapeutic drugs cannot meet the clinical demand. Herein, this work has investigated the key redox couple involved in AKI and develops a tailored photoacoustic (PA) imaging probe (AB-DiOH) which can reversibly respond to hypochlorite (ClO-)/glutathione (GSH) with high specificity and sensitivity. This probe enables the real-time monitoring of AKI by noninvasive PA imaging, with better detection sensitivity than the blood test. Furthermore, this probe is utilized for screening nephroprotective drugs among natural products. For the first time, astragalin is discovered to be a potential new drug for the treatment of AKI. After oral administration, astragalin can be efficiently absorbed by the animal body, alleviate kidney injury, and meanwhile induce no damage to other normal tissues. The treatment mechanism of astragalin has also been revealed to be the simultaneous inhibition of oxidative stress, ferroptosis, and cuproposis. The developed PA imaging probe and the discovered drug candidate provide a promising new tool and strategy for the early diagnosis and effective treatment of AKI.
Collapse
Affiliation(s)
- Wangning Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Chenming Chan
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Kaiyu Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Haifeng Qin
- Guangdong Provincial Key Laboratory of Nanophotonic Manipulation, Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Bo-Yang Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhaoli Xue
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Xianchuang Zheng
- Guangdong Provincial Key Laboratory of Nanophotonic Manipulation, Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Jiangwei Tian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
13
|
Feng W, Zhu N, Xia Y, Huang Z, Hu J, Guo Z, Li Y, Zhou S, Liu Y, Liu D. Melanin-like nanoparticles alleviate ischemia-reperfusion injury in the kidney by scavenging reactive oxygen species and inhibiting ferroptosis. iScience 2024; 27:109504. [PMID: 38632989 PMCID: PMC11022057 DOI: 10.1016/j.isci.2024.109504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/13/2024] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Kidney transplantation is essential for patients with end-stage renal disease; however, ischemia-reperfusion injury (IRI) during transplantation can lead to acute kidney damage and compromise survival. Recent studies have reported that antiferroptotic agents may be a potential therapeutic strategy, by reducing production of reactive oxygen species (ROS). Therefore, we constructed rutin-loaded polydopamine nanoparticles (PEG-PDA@rutin NPs, referred to as PPR NPs) to eliminate ROS resulting from IRI. Physicochemical characterization showed that the PPR NPs were ∼100 nm spherical particles with good ROS scavenging ability. Notably, PPR NPs could effectively enter lipopolysaccharide (LPS)-treated renal tubular cells, then polydopamine (PDA) released rutin to eliminate ROS, repair mitochondria, and suppress ferroptosis. Furthermore, in vivo imaging revealed that PPR NPs efficiently accumulated in the kidneys after IRI and effectively protected against IRI damage. In conclusion, PPR NPs demonstrated an excellent ability to eliminate ROS, suppress ferroptosis, and protect kidneys from IRI.
Collapse
Affiliation(s)
- Wenxiang Feng
- Department of Organ Transplantation, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Nan Zhu
- Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yubin Xia
- Department of Nephrology, First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Rd, Shantou, Guangdong Province 515000, China
| | - Zehai Huang
- Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jianmin Hu
- Department of Organ Transplantation, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zefeng Guo
- Department of Organ Transplantation, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yuzhuz Li
- Department of Organ Transplantation, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Song Zhou
- Department of Organ Transplantation, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yongguang Liu
- Department of Organ Transplantation, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ding Liu
- Department of Organ Transplantation, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
14
|
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
|
15
|
Guo H, Lan T, Lu X, Geng K, Shen X, Mao H, Guo Q. ROS-responsive curcumin-encapsulated nanoparticles for AKI therapy via promoting lipid degradation in renal tubules. J Mater Chem B 2024; 12:3063-3078. [PMID: 38441636 DOI: 10.1039/d3tb02318d] [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: 03/21/2024]
Abstract
Lipid accumulation is a factor contributing to the pathogenesis of acute kidney injury (AKI), yet there are currently no approved pharmacotherapies aside from adjuvant therapy. A developed reactive oxygen species (ROS)-responsive drug delivery system (NPSBG@Cur) was developed to deliver the autophagy activator curcumin (Cur) in order to alleviate AKI by activating autophagy and promoting lipid droplet degradation. The nanoparticles were shown to be ROS-responsive in the H2O2 medium and demonstrate ROS-responsive uptake in palmitate (PA)-induced oxidative stress-damaged cells. NPSBG@Cur was found to effectively inhibit lipid accumulation by autophagosome transport in kidney tubular cells. Additionally, in a mouse AKI model, NPSBG@Cur was observed to significantly ameliorate renal damage by activating autophagy flux and improving lipid transport. These results suggest that the ROS-responsive drug delivery system augmented the therapeutic effect of Cur on AKI by improving lipid metabolism through autophagy activation. Therefore, targeting lipid metabolism with NPSBG@Cur may be a promising AKI treatment strategy.
Collapse
Affiliation(s)
- Honglei Guo
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing 210029, China.
| | - Tianyu Lan
- College of Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, Guizhou Province, China.
| | - Xin Lu
- The State Key Laboratory of Functions and Applications of MediEucal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 550025, China.
| | - Kedui Geng
- The State Key Laboratory of Functions and Applications of MediEucal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 550025, China.
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of MediEucal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 550025, China.
| | - Huijuan Mao
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing 210029, China.
| | - Qianqian Guo
- The State Key Laboratory of Functions and Applications of MediEucal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 550025, China.
| |
Collapse
|
16
|
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
|
17
|
Xu T, Ning X, Wu J, Wang Q, Wang Z, Chen Z, Tang X, Bai P, Pu K, Li L, Zhang R. Metabolic Nanoregulator Remodels Gut Microenvironment for Treatment of Inflammatory Bowel Disease. ACS NANO 2024; 18:7123-7135. [PMID: 38390866 DOI: 10.1021/acsnano.3c11496] [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: 02/24/2024]
Abstract
Inflammatory bowel disease (IBD) is strongly related to the occurrence of accumulation of toxic reactive oxygen species (ROS), inflammation of the mucosa, and an imbalance of intestinal microbes. However, current treatments largely focus on a single factor, yielding unsatisfactory clinical outcomes. Herein, we report a biocompatible and IBD-targeted metabolic nanoregulator (TMNR) that synergistically regulates cellular and bacterial metabolism. The TMNR comprises a melanin-gallium complex (MNR) encapsulated within a thermosensitive and colitis-targeting hydrogel, all composed of natural and FDA-approved components. The TMNR confers superior broad-spectrum antioxidant properties, effectively scavenging reactive oxygen species (ROS) and blocking inflammatory signaling pathways. The presence of Ga3+ in TMNR selectively disrupts iron metabolism in pathogenic microorganisms due to its structural resemblance to the iron atom. Additionally, incorporating a thermosensitive injectable hydrogel enables targeted delivery of TMNR to inflammatory regions, prolonging their retention time and providing a physical barrier function for optimizing IBD treatment efficacy. Collectively, TMNR effectively modulates the redox balance of inflamed colonic epithelial tissue and disrupts iron metabolism in pathogenic microorganisms, thereby eliminating inflammation and restoring intestinal homeostasis against IBD. Hence, this work presents a comprehensive approach for precise spatiotemporal regulation of the intestinal microenvironmental metabolism for IBD treatment.
Collapse
Affiliation(s)
- Ting Xu
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Xiaogang Ning
- School of Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Jiayan Wu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 637457, Singapore
| | - Qian Wang
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Zhifei Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Zhiqing Chen
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Xiaoxian Tang
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Peirong Bai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 637457, Singapore
| | - Liping Li
- The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| |
Collapse
|
18
|
Bao X, Huo S, Wang Z, Yang S, Dou L, Liu Y, Huang J, Cai C, Fang B, Xu G. Multifunctional biomimetic hydrogel dressing provides anti-infection treatment and improves immunotherapy by reprogramming the infection-related wound microenvironment. J Nanobiotechnology 2024; 22:80. [PMID: 38418972 PMCID: PMC10902999 DOI: 10.1186/s12951-024-02337-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024] Open
Abstract
The advancement of biomaterials with antimicrobial and wound healing properties continues to present challenges. Macrophages are recognized for their significant role in the repair of infection-related wounds. However, the interaction between biomaterials and macrophages remains complex and requires further investigation. In this research, we propose a new sequential immunomodulation method to enhance and expedite wound healing by leveraging the immune properties of bacteria-related wounds, utilizing a novel mixed hydrogel dressing. The hydrogel matrix is derived from porcine acellular dermal matrix (PADM) and is loaded with a new type of bioactive glass nanoparticles (MBG) doped with magnesium (Mg-MBG) and loaded with Curcumin (Cur). This hybrid hydrogel demonstrates controlled release of Cur, effectively eradicating bacterial infection in the early stage of wound infection, and the subsequent release of Mg ions (Mg2+) synergistically inhibits the activation of inflammation-related pathways (such as MAPK pathway, NF-κB pathway, TNF-α pathway, etc.), suppressing the inflammatory response caused by infection. Therefore, this innovative hydrogel can safely and effectively expedite wound healing during infection. Our design strategy explores novel immunomodulatory biomaterials, offering a fresh approach to tackle current clinical challenges associated with wound infection treatment.
Collapse
Affiliation(s)
- Xiaogang Bao
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Shicheng Huo
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| | - Zhenhua Wang
- Department of Laboratory Medicine, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Shengyan Yang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Linyun Dou
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Yifei Liu
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Jian Huang
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Chang Cai
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Bin Fang
- Department of Orthopedics, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, China.
| | - Guohua Xu
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| |
Collapse
|
19
|
Zou Q, Chen X, Li B, Zhang R, Pan J, Zhang X, Zhang X, Sun SK. Bioinspired BSA@polydopamine@Fe Nanoprobe with Self-Purification Capacity for Targeted Magnetic Resonance Imaging of Acute Kidney Injury. ACS NANO 2024; 18:4783-4795. [PMID: 38301134 DOI: 10.1021/acsnano.3c09193] [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: 02/03/2024]
Abstract
Contrast-enhanced magnetic resonance imaging (CE-MRI) of acute kidney injury (AKI) is severely hindered by the poor targeting capacity and potential toxicity of current contrast agents. Herein, we propose one-step fabrication of a bovine serum albumin@polydopamine@Fe (BSA@PDA@Fe, BPFe) nanoprobe with self-purification capacity for targeted CE-MRI of AKI. BSA endows the BPFe nanoprobe with renal tubule-targeting ability, and PDA is capable of completely inhibiting the intrinsic metal-induced reactive oxygen species (ROS), which are always involved in Fe/Mn-based agents. The as-prepared nanoprobe owns a tiny size of 2.7 nm, excellent solubility, good T1 MRI ability, superior biocompatibility, and powerful antioxidant capacity. In vivo CE-MRI shows that the BPFe nanoprobe can accumulate in the renal cortex due to the reabsorption effect toward the serum albumin. In the AKI model, impaired renal reabsorption function can be effortlessly detected via the diminishment of renal cortical signal enhancement. More importantly, the administration of the BPFe nanoprobe would not aggravate renal damage of AKI due to the outstanding self-purification capacity. Besides, the BPFe nanoprobe is employed for CE-MR angiography to visualize fine vessel structures. This work provides an MRI contrast agent with good biosafety and targeting ability for CE-MRI of kidney diseases.
Collapse
Affiliation(s)
- Quan Zou
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xi Chen
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China
| | - Bingjie Li
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ruijie Zhang
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China
| | - Jinbin Pan
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xuejun Zhang
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China
| | - Xuening Zhang
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300203, China
| |
Collapse
|
20
|
Zhou L, Yang Z, Guo L, Zou Q, Zhang H, Sun SK, Ye Z, Zhang C. Noninvasive Assessment of Kidney Injury by Combining Structure and Function Using Artificial Intelligence-Based Manganese-Enhanced Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5474-5485. [PMID: 38271189 DOI: 10.1021/acsami.3c14936] [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: 01/27/2024]
Abstract
Contrast-enhanced magnetic resonance imaging (MRI) is seriously limited in kidney injury detection due to the nephrotoxicity of clinically used gadolinium-based contrast agents. Herein, we propose a noninvasive method for the assessment of kidney injury by combining structure and function information based on manganese (Mn)-enhanced MRI for the first time. As a proof of concept, the Mn-melanin nanoprobe with good biocompatibility and excellent T1 relaxivity is applied in MRI of a unilateral ureteral obstruction mice model. The abundant renal structure and function information is obtained through qualitative and quantitative analysis of MR images, and a brand new comprehensive assessment framework is proposed to precisely identify the degree of kidney injury successfully. Our study demonstrates that Mn-enhanced MRI is a promising approach for the highly sensitive and biosafe assessment of kidney injury in vivo.
Collapse
Affiliation(s)
- Li Zhou
- Department of Radiology, Tianjin Chest Hospital, Tianjin 300052, China
| | - Zizhen Yang
- Department of Radiology, Ningbo No.2 Hospital, Ningbo 315012, China
| | - Li Guo
- School of Medical Imaging, Tianjin Medical University, Tianjin 300203, China
| | - Quan Zou
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Hong Zhang
- Department of Radiology, Tianjin Chest Hospital, Tianjin 300052, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Medical University, Tianjin 300203, China
| | - Zhaoxiang Ye
- Department of Radiology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Cai Zhang
- Department of Radiology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
21
|
Wang Y, Lu Z, Liu B, Seidi F, Zhang C, Jiang B, Huang C, Xiao H, Wang P, Jin Y. Antitumor Effects of Carrier-Free Functionalized Lignin Materials on Human Hepatocellular Carcinoma (HepG2) Cells. ACS NANO 2024; 18:4329-4342. [PMID: 38261787 DOI: 10.1021/acsnano.3c09924] [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: 01/25/2024]
Abstract
Lignin, as an abundant aromatic biopolymer in plants, has great potential for medical applications due to its active sites, antioxidant activity, low biotoxicity, and good biocompatibility. In this work, a simple and ecofriendly approach for lignin fractionation and modification was developed to improve the antitumor activity of lignin. The lignin fraction KL-3 obtained by the lignin gradient acid precipitation at pH = 9-13 showed good cytotoxicity. Furthermore, the cell-feeding lignin after additional structural modifications such as demethylation (DKL-3), sulfonation (SL-3), and demethylsulfonation (DSKL-3) could exhibit higher glutathione responsiveness in the tumor microenvironment, resulting in reactive oxygen species accumulation and mitochondrial damage and eventually leading to apoptosis in HepG2 cells with minimal damage to normal cells. The IC50 values for KL-3, SL-3, and DSKL-3 were 0.71, 0.57, and 0.41 mg/mL, respectively, which were superior to those of other biomass extractives or unmodified lignin. Importantly, in vivo experiments conducted in nude mouse models demonstrated good biosafety and effective tumor destruction. This work provides a promising example of constructing carrier-free functionalized lignin antitumor materials with different structures for inhibiting the growth of human hepatocellular carcinoma (HepG2) cells, which is expected to improve cancer therapy outcomes.
Collapse
Affiliation(s)
- Yilin Wang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Zhiqiang Lu
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Bin Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Farzad Seidi
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Chaofeng Zhang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Bo Jiang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Caoxing Huang
- Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Huining Xiao
- Department of Chemical Engineering, University of New Brunswick, Fredericton E3B 5A3, Canada
| | - Peng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yongcan Jin
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
22
|
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
|
23
|
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: 1.0] [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
|
24
|
Sun J, Han Y, Dong J, Lv S, Zhang R. Melanin/melanin-like nanoparticles: As a naturally active platform for imaging-guided disease therapy. Mater Today Bio 2023; 23:100894. [PMID: 38161509 PMCID: PMC10755544 DOI: 10.1016/j.mtbio.2023.100894] [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: 10/04/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
The development of biocompatible and efficient nanoplatforms that combine diagnostic and therapeutic functions is of great importance for precise disease treatment. Melanin, an endogenous biopolymer present in living organisms, has attracted increasing attention as a versatile bioinspired functional platform owing to its unique physicochemical properties (e.g., high biocompatibility, strong chelation of metal ions, broadband light absorption, high drug binding properties) and inherent antioxidant, photoprotective, anti-inflammatory, and anti-tumor effects. In this review, the fundamental physicochemical properties and preparation methods of natural melanin and melanin-like nanoparticles were outlined. A systematical description of the recent progress of melanin and melanin-like nanoparticles in single, dual-, and tri-multimodal imaging-guided the visual administration and treatment of osteoarthritis, acute liver injury, acute kidney injury, acute lung injury, brain injury, periodontitis, iron overload, etc. Was then given. Finally, it concluded with a reasoned discussion of current challenges toward clinical translation and future striving directions. Therefore, this comprehensive review provides insight into the current status of melanin and melanin-like nanoparticles research and is expected to optimize the design of novel melanin-based therapeutic platforms and further clinical translation.
Collapse
Affiliation(s)
- Jinghua Sun
- The Molecular Medicine Research Team of First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Yahong Han
- Shanxi Medical University, Taiyuan 030001, China
| | - Jie Dong
- Shanxi Medical University, Taiyuan 030001, China
| | - Shuxin Lv
- Shanxi Medical University, Taiyuan 030001, China
| | - Ruiping Zhang
- The Molecular Medicine Research Team of First Hospital of Shanxi Medical University, Taiyuan, 030001, China
- The Radiology Department of Shanxi Provincial People’ Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
25
|
He S, Chen C, Li F, Xu W, Li D, Liang M, Yang X. A Polymeric Nanosponge as a Broad-Spectrum Reactive Oxygen Species Scavenger for Acute Kidney Injury Treatment. NANO LETTERS 2023; 23:8978-8987. [PMID: 37726233 DOI: 10.1021/acs.nanolett.3c02531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Acute kidney injury (AKI) is closely associated with the overproduction of reactive oxygen species (ROS), which can cause multiple organ dysfunctions without timely treatment. However, only supportive treatments are currently available for AKI in clinics. Here, we developed nanomaterials of hyperbranched polyphosphoester (PPE) containing abundant thioether (S-PPE NP) and thioketal bonds (TK-PPE NP). Our data demonstrates that S-PPE NP exhibits an excellent capability of absorbing and scavenging multiple types of ROS, including H2O2, •OH, and •O2-, via thioether oxidation to sulfone or sulfoxide; it was also determined that S-PPE NP efficiently eliminates intracellular ROS, thus preventing cellular damage. Moreover, S-PPE NP was able to efficiently accumulate in the injured kidneys of AKI-bearing mice. As a result, the administration of S-PPE NP provided a superior therapeutic effect in AKI-bearing mice by downregulating ROS- and inflammation-related signaling pathways, thus reducing cell apoptosis. This thioether-containing polymer represents a promising broad-spectrum ROS scavenger that can be used for effective AKI treatments.
Collapse
Affiliation(s)
- Shan He
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
| | - Chaoran Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
| | - Fangzheng Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
| | - Wenxuan Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
| | - Dongdong Li
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
| | - Ming Liang
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
| | - Xianzhu Yang
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, P. R. China
| |
Collapse
|
26
|
Zhou X, Wang N, Liu W, Chen R, Yang G, Yu H. Identification of the potential association between SARS-CoV-2 infection and acute kidney injury based on the shared gene signatures and regulatory network. BMC Infect Dis 2023; 23:655. [PMID: 37789254 PMCID: PMC10548629 DOI: 10.1186/s12879-023-08638-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/24/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is identified as the cause of coronavirus disease 2019 (COVID-19) pandemic. Acute kidney injury (AKI), one of serious complications of COVID-19 infection, is the leading contributor to renal failure, associating with high mortality of the patients. This study aimed to identify the shared gene signatures and construct the gene regulatory network between COVID-19 and AKI, contributing to exploring the potential pathogenesis. METHODS Utilizing the machine learning approach, the candidate gene signatures were derived from the common differentially expressed genes (DEGs) obtained from COVID-19 and AKI. Subsequently, receiver operating characteristic (ROC), consensus clustering and functional enrichment analyses were performed. Finally, protein-protein interaction (PPI) network, transcription factor (TF)-gene interaction, gene-miRNA interaction, and TF-miRNA coregulatory network were systematically undertaken. RESULTS We successfully identified the shared 6 candidate gene signatures (RRM2, EGF, TMEM252, RARRES1, COL6A3, CUBN) between COVID-19 and AKI. ROC analysis showed that the model constructed by 6 gene signatures had a high predictive efficacy in COVID-19 (AUC = 0.965) and AKI (AUC = 0.962) cohorts, which had the potential to be the shared diagnostic biomarkers for COVID-19 and AKI. Additionally, the comprehensive gene regulatory networks, including PPI, TF-gene interaction, gene-miRNA interaction, and TF-miRNA coregulatory networks were displayed utilizing NetworkAnalyst platform. CONCLUSIONS This study successfully identified the shared gene signatures and constructed the comprehensive gene regulatory network between COVID-19 and AKI, which contributed to predicting patients' prognosis and providing new ideas for developing therapeutic targets for COVID-19 and AKI.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Nephrology, Haihe Hospital, Tianjin University, 890 Jingu Road, Jinnan District, Tianjin, 300350, China.
- Department of Nephrology, Tianjin Haihe Hospital, Tianjin, 300350, China.
- Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China.
- Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China.
| | - Ning Wang
- The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
| | - Wenjing Liu
- Department of Nephrology, Tianjin Haihe Hospital, Tianjin, 300350, China
| | - Ruixue Chen
- Tianjin Haihe Hospital, Tianjin, 300350, China
| | - Guoyue Yang
- The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China.
| | - Hongzhi Yu
- Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China.
- Department of Respiratory Medicine, Tianjin Haihe Hospital, 890 Jingu Road, Jinnan District, Tianjin, 300350, China.
| |
Collapse
|
27
|
Geng S, Feng Q, Wang C, Li Y, Qin J, Hou M, Zhou J, Pan X, Xu F, Fang B, Wang K, Yu Z. A Versatile PDA(DOX) Nanoplatform for Chemo-Photothermal Synergistic Therapy against Breast Cancer and Attenuated Doxorubicin-Induced Cardiotoxicity. J Nanobiotechnology 2023; 21:338. [PMID: 37735669 PMCID: PMC10512561 DOI: 10.1186/s12951-023-02072-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
Photothermal therapy (PTT) is a highly clinical application promising cancer treatment strategy with safe, convenient surgical procedures and excellent therapeutic efficacy on superficial tumors. However, a single PTT is difficult to eliminate tumor cells completely, and tumor recurrence and metastasis are prone to occur in the later stage. Chemo-photothermal synergistic therapy can conquer the shortcomings by further killing residual tumor cells after PTT through systemic chemotherapy. Nevertheless, chemotherapy drugs' extreme toxicity is also a problematic issue to be solved, such as anthracycline-induced cardiotoxicity. Herein, we selected polydopamine nanoparticles (PDA) as the carrier of the chemotherapeutic drug doxorubicin (DOX) to construct a versatile PDA(DOX) nanoplatform for chemo-photothermal synergistic therapy against breast cancer and simultaneously attenuated DOX-induced cardiotoxicity (DIC). The excellent photothermal properties of PDA were used to achieve the thermal ablation of tumors. DOX carried out chemotherapy to kill residual and occult distant tumors. Furthermore, the PDA(DOX) nanoparticles significantly alleviate DIC, which benefits from PDA's excellent antioxidant enzyme activity. The experimental data of the chemotherapy groups showed that the results of the PDA(DOX) group were much better than the DOX group. This study not only effectively inhibits cancer but tactfully attenuates DIC, bringing a new perspective into synergistic therapy against breast cancer.
Collapse
Affiliation(s)
- Siqi Geng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Qiang Feng
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Chujie Wang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Ying Li
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Jiaying Qin
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Mingsheng Hou
- Department of Pathology, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Jiedong Zhou
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Xiaoyu Pan
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Fei Xu
- Department of Ultrasound, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Baoru Fang
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Ke Wang
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China
| | - Zhangsen Yu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China.
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, People's Republic of China.
| |
Collapse
|
28
|
Tian Y, Zhang Y, Zhao J, Luan F, Wang Y, Lai F, Ouyang D, Tao Y. Combining MSC Exosomes and Cerium Oxide Nanocrystals for Enhanced Dry Eye Syndrome Therapy. Pharmaceutics 2023; 15:2301. [PMID: 37765270 PMCID: PMC10536361 DOI: 10.3390/pharmaceutics15092301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Dry eye syndrome (DES) is a prevalent ocular disorder involving diminishe·d tear production and increased tear evaporation, leading to ocular discomfort and potential surface damage. Inflammation and reactive oxygen species (ROS) have been implicated in the pathophysiology of DES. Inflammation is one core cause of the DES vicious cycle. Moreover, there are ROS that regulate inflammation in the cycle from the upstream, which leads to treatment failure in current therapies that merely target inflammation. In this study, we developed a novel therapeutic nanoparticle approach by growing cerium oxide (Ce) nanocrystals in situ on mesenchymal stem cell-derived exosomes (MSCExos), creating MSCExo-Ce. The combined properties of MSCExos and cerium oxide nanocrystals aim to target the "inflammation-ROS-injury" pathological mechanism in DES. We hypothesized that this approach would provide a new treatment option for patients with DES. Our analysis confirmed the successful in situ crystallization of cerium onto MSCExos, and MSCExo-Ce displayed excellent biocompatibility. In vitro and in vivo experiments have demonstrated that MSCExo-Ce promotes corneal cell growth, scavenges ROS, and more effectively suppresses inflammation compared with MSCExos alone. MSCExo-Ce also demonstrated the ability to alleviate DES symptoms and reverse pathological alterations at both the cellular and tissue levels. In conclusion, our findings highlight the potential of MSCExo-Ce as a promising therapeutic candidate for the treatment of DES.
Collapse
Affiliation(s)
- Ying Tian
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yiquan Zhang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jiawei Zhao
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Fuxiao Luan
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yingjie Wang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Fan Lai
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, Center for Life Science, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau 999078, China
| | - Yong Tao
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
29
|
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: 3] [Impact Index Per Article: 3.0] [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
|
30
|
Ding C, Wang B, Zheng J, Zhang M, Li Y, Shen HH, Guo Y, Zheng B, Tian P, Ding X, Xue W. Neutrophil Membrane-Inspired Nanorobots Act as Antioxidants Ameliorate Ischemia Reperfusion-Induced Acute Kidney Injury. ACS APPLIED MATERIALS & INTERFACES 2023; 15:40292-40303. [PMID: 37603686 DOI: 10.1021/acsami.3c08573] [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: 08/23/2023]
Abstract
Ischemia/reperfusion (I/R) injury causes excessive oxidative events and initiates destructive inflammatory responses, and it is an important promoter to the pathology of various pathema states. Ferroptosis is an iron-dependent type of nonapoptotic cell death accompanied by the accumulation of membrane lipid peroxide and consumption of polyunsaturated fatty acid, and it plays a key role in I/R injury diseases. Moreover, the excessive production of inflammatory cytokines contributes to the development of acute kidney injury. Here, we reported neutrophil membrane-coated copper-based nanoparticles (N-Cu5.4O@DFO NPs) for I/R kidney injury treatment. The highly biocompatible and stable N-Cu5.4O@DFO NPs showed excellent antioxidant and iron ion scavenging abilities in vitro. Our finding showed that the N-Cu5.4O@DFO NPs strategy could significantly accumulate in the inflammatory kidney, reduce oxidative damage events and inflammatory response, and finally achieve synergistic therapy against renal I/R injury. This work promotes the development of nanoantioxidant agents with multiple antioxidant properties for the therapy of other I/R injury diseases.
Collapse
Affiliation(s)
- Chenguang Ding
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an 710061, China
- Organ Procurement and Allocation Organization, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Bo Wang
- Department of Material Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Zheng
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an 710061, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yang Li
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an 710061, China
| | - Hsin-Hui Shen
- Department of Material Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Yingcong Guo
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Bingxuan Zheng
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Puxun Tian
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoming Ding
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an 710061, China
| | - Wujun Xue
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
31
|
Xu Z, Zhu Y, Xie M, Liu K, Cai L, Wang H, Li D, Chen H, Gao L. Mackinawite nanozymes as reactive oxygen species scavengers for acute kidney injury alleviation. J Nanobiotechnology 2023; 21:281. [PMID: 37598162 PMCID: PMC10439570 DOI: 10.1186/s12951-023-02034-7] [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: 04/26/2023] [Accepted: 07/31/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Iron sulfide nanomaterials have been successfully employed as therapeutic agents for bacterial infection therapy and catalytic-ferroptosis synergistic tumor therapy due to their unique structures, physiochemical properties, and biocompatibility. However, biomedical research and understanding of the biological functions of iron sulfides are insufficient, and how iron sulfide nanomaterials affect reactive oxygen species (ROS) in diseases remains unknown. Acute kidney injury (AKI) is associated with high levels of ROS, and therefore nanomedicine-mediated antioxidant therapy has emerged as a novel strategy for its alleviation. RESULTS Here, mackinawite nanozymes were synthesized from glutathione (GSH) and iron ions (Fe3+) (denoted as GFeSNs) using a hydrothermal method, and then evaluated as ROS scavengers for ROS-related AKI treatment. GFeSNs showed broad-spectrum ROS scavenging ability through synergistic interactions of multiple enzymes-like and hydrogen polysulfide-releasing properties. Furthermore, both in vitro and in vivo experiments demonstrated that GFeSNs exhibited outstanding cytoprotective effects against ROS-induced damage at extremely low doses and significantly improved treatment outcomes in AKI. CONCLUSIONS Given the synergetic antioxidant properties and high biocompatibility, GFeSNs exhibit great potential for the treatment of AKI and other ROS-associated diseases.
Collapse
Affiliation(s)
- Zhuobin Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Yufei Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Mengke Xie
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Kankan Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Liangliang Cai
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Huihui Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Dandan Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| | - Hao Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
32
|
Wu J, Shang H, Zhang A, He Y, Tong Y, Huang Q, Liu X, Chen Z, Tang K. Antioxidant nanozymes in kidney injury: mechanism and application. NANOSCALE 2023; 15:13148-13171. [PMID: 37547960 DOI: 10.1039/d3nr01954c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Excessive production of reactive oxygen species (ROS) in the kidneys is involved in the pathogenesis of kidney diseases, such as acute kidney injury (AKI) and diabetic kidney disease (DKD), and is the main reason for the progression of kidney injury. ROS can easily lead to lipid peroxidation and damage the tubular epithelial cell membrane, proteins and DNA, and other molecules, which can trigger cellular oxidative stress. Effective scavenging of ROS can delay or halt the progression of kidney injury by reducing inflammation and oxidative stress. With the development of nanotechnology and an improved understanding of nanomaterials, more researchers are applying nanomaterials with antioxidant activity to treat kidney injury. This article reviews the detailed mechanism between ROS and kidney injury, as well as the applications of nanozymes with antioxidant effects based on different materials for various kidney injuries. To better guide the applications of antioxidant nanozymes in kidney injury and other inflammatory diseases, at the end of this review we also summarize the aspects of nanozymes that need to be improved. An in-depth understanding of the role played by ROS in the occurrence and progression of kidney injury and the mechanism by which antioxidant nanozymes reduce oxidative stress is conducive to improving the therapeutic effect in kidney injury and inflammation-related diseases.
Collapse
Affiliation(s)
- Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - An Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Qiaokou District, Wuhan, 430030, China.
| |
Collapse
|
33
|
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: 23] [Impact Index Per Article: 23.0] [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
|
34
|
Morilla MJ, Ghosal K, Romero EL. More Than Pigments: The Potential of Astaxanthin and Bacterioruberin-Based Nanomedicines. Pharmaceutics 2023; 15:1828. [PMID: 37514016 PMCID: PMC10385456 DOI: 10.3390/pharmaceutics15071828] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Carotenoids are natural products regulated by the food sector, currently used as feed dyes and as antioxidants in dietary supplements and composing functional foods for human consumption. Of the nearly one thousand carotenoids described to date, only retinoids, derived from beta carotene, have the status of a drug and are regulated by the pharmaceutical sector. In this review, we address a novel field: the transformation of xanthophylls, particularly the highly marketed astaxanthin and the practically unknown bacterioruberin, in therapeutic agents by altering their pharmacokinetics, biodistribution, and pharmacodynamics through their formulation as nanomedicines. The antioxidant activity of xanthophylls is mediated by routes different from those of the classical oral anti-inflammatory drugs such as corticosteroids and non-steroidal anti-inflammatory drugs (NSAIDs): remarkably, xanthophylls lack therapeutic activity but also lack toxicity. Formulated as nanomedicines, xanthophylls gain therapeutic activity by mechanisms other than increased bioavailability. Loaded into ad hoc tailored nanoparticles to protect their structure throughout storage and during gastrointestinal transit or skin penetration, xanthophylls can be targeted and delivered to selected inflamed cell groups, achieving a massive intracellular concentration after endocytosis of small doses of formulation. Most first reports showing the activities of oral and topical anti-inflammatory xanthophyll-based nanomedicines against chronic diseases such as inflammatory bowel disease, psoriasis, atopic dermatitis, and dry eye disease emerged between 2020 and 2023. Here we discuss in detail their preclinical performance, mostly targeted vesicular and polymeric nanoparticles, on cellular models and in vivo. The results, although preliminary, are auspicious enough to speculate upon their potential use for oral or topical administration in the treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Maria Jose Morilla
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Saenz Peña 352, Bernal 1876, Argentina
| | - Kajal Ghosal
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja Subodh Chandra Mallick Rd., Jadavpur, Kolkata 700032, West Bengal, India
| | - Eder Lilia Romero
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Saenz Peña 352, Bernal 1876, Argentina
| |
Collapse
|
35
|
Mavridi-Printezi A, Menichetti A, Mordini D, Montalti M. Functionalization of and through Melanin: Strategies and Bio-Applications. Int J Mol Sci 2023; 24:9689. [PMID: 37298641 PMCID: PMC10253489 DOI: 10.3390/ijms24119689] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
A unique feature of nanoparticles for bio-application is the ease of achieving multi-functionality through covalent and non-covalent functionalization. In this way, multiple therapeutic actions, including chemical, photothermal and photodynamic activity, can be combined with different bio-imaging modalities, such as magnetic resonance, photoacoustic, and fluorescence imaging, in a theragnostic approach. In this context, melanin-related nanomaterials possess unique features since they are intrinsically biocompatible and, due to their optical and electronic properties, are themselves very efficient photothermal agents, efficient antioxidants, and photoacoustic contrast agents. Moreover, these materials present a unique versatility of functionalization, which makes them ideal for the design of multifunctional platforms for nanomedicine integrating new functions such as drug delivery and controlled release, gene therapy, or contrast ability in magnetic resonance and fluorescence imaging. In this review, the most relevant and recent examples of melanin-based multi-functionalized nanosystems are discussed, highlighting the different methods of functionalization and, in particular, distinguishing pre-functionalization and post-functionalization. In the meantime, the properties of melanin coatings employable for the functionalization of a variety of material substrates are also briefly introduced, especially in order to explain the origin of the versatility of melanin functionalization. In the final part, the most relevant critical issues related to melanin functionalization that may arise during the design of multifunctional melanin-like nanoplatforms for nanomedicine and bio-application are listed and discussed.
Collapse
Affiliation(s)
| | | | | | - Marco Montalti
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy; (A.M.-P.); (A.M.); (D.M.)
| |
Collapse
|
36
|
Bao B, Liu H, Han Y, Xu L, Xing W, Li Z. Simultaneous Elimination of Reactive Oxygen Species and Activation of Nrf2 by Ultrasmall Nanoparticles to Relieve Acute Kidney Injury. ACS APPLIED MATERIALS & INTERFACES 2023; 15:16460-16470. [PMID: 36946292 DOI: 10.1021/acsami.3c00052] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Excess reactive oxygen species (ROS) can induce serious acute kidney injury (AKI) to result in numerous deaths annually in clinical practice. Elimination of excess ROS by advanced nanotechnology is a very promising AKI therapy. In this Article, we report that PVP-stabilized and quercetin-functionalized ultrasmall Cu2-xSe nanoparticles (abbreviated as CSPQ NPs) can efficiently scavenge ROS and increase the expression of intracellular antioxidative enzymes by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) protein, which drastically alleviates the cellular oxidative stress. Our ultrasmall nanoparticles exhibit excellent biocompatibility. They can be rapidly accumulated into the injured kidney to simultaneously eliminate ROS and activate Nrf2 to improve the renal function. This work demonstrates the great potential of simultaneous elimination of ROS and activation of intracellular Nrf2 in treatment of AKI. It also highlights the potential of CSPQ NPs in protection and prevention of AKI.
Collapse
Affiliation(s)
- Bolin Bao
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P. R. China
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China
| | - Hanghang Liu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China
| | - Liyao Xu
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P. R. China
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China
| | - Wei Xing
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P. R. China
| | - Zhen Li
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P. R. China
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, P. R. China
| |
Collapse
|
37
|
Mavridi-Printezi A, Menichetti A, Mordini D, Amorati R, Montalti M. Recent Applications of Melanin-like Nanoparticles as Antioxidant Agents. Antioxidants (Basel) 2023; 12:antiox12040863. [PMID: 37107238 PMCID: PMC10135245 DOI: 10.3390/antiox12040863] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Nanosized antioxidants are highly advantageous in terms of versatility and pharmacokinetics, with respect to conventional molecular ones. Melanin-like materials, artificial species inspired by natural melanin, combine recognized antioxidant (AOX) activity with a unique versatility of preparation and modification. Due to this versatility and documented biocompatibility, artificial melanin has been incorporated into a variety of nanoparticles (NP) in order to give new platforms for nanomedicine with enhanced AOX activity. In this review article, we first discuss the chemical mechanisms behind the AOX activity of materials in the context of the inhibition of the radical chain reaction responsible for the peroxidation of biomolecules. We also focus briefly on the AOX properties of melanin-like NP, considering the effect of parameters such as size, preparation methods and surface functionalization on them. Then, we consider the most recent and relevant applications of AOX melanin-like NPs that are able to counteract ferroptosis and be involved in the treatment of important diseases that affect, e.g., the cardiovascular and nervous systems, as well as the kidneys, liver and articulations. A specific section will be dedicated to cancer treatment, since the role of melanin in this context is still very debated. Finally, we propose future strategies in AOX development for a better chemical understanding of melanin-like materials. In particular, the composition and structure of these materials are still debated, and they present a high level of variability. Thus, a better understanding of the mechanism behind the interaction of melanin-like nanostructures with different radicals and highly reactive species would be highly advantageous for the design of more effective and specific AOX nano-agents.
Collapse
Affiliation(s)
| | - Arianna Menichetti
- Department of Chemistry «Giacomo Ciamician», University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Dario Mordini
- Department of Chemistry «Giacomo Ciamician», University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Riccardo Amorati
- Department of Chemistry «Giacomo Ciamician», University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Marco Montalti
- Department of Chemistry «Giacomo Ciamician», University of Bologna, Via Selmi 2, 40126 Bologna, Italy
- Tecnopolo di Rimini, Via Dario Campana 71, 47921 Rimini, Italy
| |
Collapse
|
38
|
Li T, Jing W, Fu W, Yan Z, Ma Y, Li X, Ji H, Zhang R. Melanin theranostic nanoplatform as an efficient drug delivery system for imaging-guided renal fibrosis therapy. BIOMATERIALS ADVANCES 2023; 147:213333. [PMID: 36801511 DOI: 10.1016/j.bioadv.2023.213333] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
As renal fibrosis nanotherapeutics, the endogenous biomaterial melanin not only has natural biocompatibility and biodegradability but also has inherent photoacoustic imaging ability and certain anti-inflammatory effects. These properties determine that melanin can not only as a carrier of medication but also track the biodistribution and renal uptake of drugs in vivo by photoacoustic imaging in real-time. Curcumin is a natural compound with biological activity, which has excellent ROS scavenging ability and good anti-inflammatory property. These materials appear more advantages in the development of nanoscale diagnostic and therapeutic platforms for future clinical translation. Herein, this study developed curcumin-loaded melanin nanoparticles (MNP-PEG-CUR NPs) as an efficient medication delivery system for photoacoustic imaging guidance renal fibrosis treatment. The nanoparticles are about 10 nm in size, exhibit good renal clearance efficiency, excellent photoacoustic imaging ability, and good in vitro and in vivo biocompatibility. These preliminary results indicated that MNP-PEG-CUR have clinically applicable potential as a therapeutic nanoplatform for renal fibrosis.
Collapse
Affiliation(s)
- Tingting Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People's Republic of China; School of Pharmacy, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Wenyu Jing
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Weihua Fu
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Zirui Yan
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yuan Ma
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Xueqi Li
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Huifang Ji
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Ruiping Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan 030001, People's Republic of China.
| |
Collapse
|
39
|
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: 38.0] [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
|
40
|
Xu L, Luo Y, Du Q, Zhang W, Hu L, Fang N, Wang J, Liu J, Zhou J, Zhong Y, Liu Y, Ran H, Guo D, Xu J. Magnetic Response Combined with Bioactive Ion Therapy: A RONS-Scavenging Theranostic Nanoplatform for Thrombolysis and Renal Ischemia-Reperfusion Injury. ACS NANO 2023; 17:5695-5712. [PMID: 36930590 DOI: 10.1021/acsnano.2c12091] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Currently, the limited efficacy of antithrombotic treatments is attributed to the inadequacy of pure drugs and the low ability of drugs to target the thrombus site. More importantly, timely thrombolysis is essential to reduce the sequelae of cardiovascular disease, but ischemia-reperfusion injury (IRI) remains a major challenge that must be solved after blood flow recovery. Herein, a multifunctional therapeutic nanoparticle (NP) based on Fe3O4 and strontium ions encapsulated in mesoporous polydopamine was successfully constructed and then loaded with TNK-tPA (FeM@Sr-TNK NPs). The NPs (59.9 min) significantly prolonged the half-life of thrombolytic drugs, which was 3.04 times that of TNK (19.7 min), and they had good biological safety. The NPs were shown to pass through vascular models with different inner diameters, curvatures, and stenosis under magnetic targeting and to enable accurate diagnosis of thrombi by photoacoustic imaging. NPs combined with the magnetic hyperthermia technique were used to accelerate thrombolysis and quickly open blocked blood vessels. Then, renal IRI-induced functional metabolic disorder and tissue damage were evidently attenuated by scavenging toxic reactive oxygen and nitrogen species and through the protective effects of bioactive ion therapy, including reduced apoptosis, increased angiogenesis, and inhibited fibrosis. In brief, we constructed a multifunctional nanoplatform for integrating a "diagnosis-therapy-protection" approach to achieve comprehensive management from thrombus to renal IRI, promoting the advancement of related technologies.
Collapse
Affiliation(s)
- Lian Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Ying Luo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Qianying Du
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Wenli Zhang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Liu Hu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Ni Fang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Junrui Wang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Jia Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Jun Zhou
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Yixin Zhong
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Yun Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Dajing Guo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Jie Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| |
Collapse
|
41
|
Yang Y, Li T, Jing W, Yan Z, Li X, Fu W, Zhang R. Dual-modality and Noninvasive Diagnostic of MNP-PEG-Mn Nanoprobe for Renal Fibrosis Based on Photoacoustic and Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12797-12808. [PMID: 36866785 DOI: 10.1021/acsami.2c22512] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
To date, imaging-guided multimodality therapy is important to improve the accuracy of the diagnosis of renal fibrosis, and nanoplatforms for imaging-guided multimodality diagnosis are gaining more and more attention. There are many limitations and deficiencies in clinical use for early-stage diagnosis of renal fibrosis, and multimodal imaging can contribute more thoroughly and provide in-detail information for effective clinical diagnosis. Melanin is an endogenous biomaterial, and we developed an ultrasmall particle size melanin nanoprobe (MNP-PEG-Mn) based on photoacoustic (PA) and magnetic resonance (MR) dual-modal imaging. MNP-PEG-Mn nanoprobe, with the average diameter about 2.7 nm, can be passively targeted for accumulation in the kidney, and it has excellent free radical scavenging and antioxidant abilities without further exacerbating renal fibrosis. Using the normal group signal as a control, the dual-modal imaging results showed that the MR imaging (MAI) and PA imaging (PAI) signals reached the strongest at 6 h when MNP-PEG-Mn entered the 7 day renal fibrosis group via the left vein of the tail end of the mice; however, the strength of the dual-modal imaging signal and the gradient of signal change were significantly weaker in the 28 day renal fibrosis group than in the 7 day renal fibrosis group and normal group. The phenomenon preliminarily indicates that as a PAI/MRI dual-modality contrast medium candidate, MNP-PEG-Mn has outstanding ability in clinical application potential.
Collapse
Affiliation(s)
- Yilin Yang
- Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Tingting Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Wenyu Jing
- Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Zirui Yan
- Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Xueqi Li
- Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Weihua Fu
- Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Ruiping Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
42
|
Biomarkers of oxidative stress and reproductive complications. Adv Clin Chem 2023; 113:157-233. [PMID: 36858646 DOI: 10.1016/bs.acc.2022.11.004] [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: 02/04/2023]
Abstract
Oxidative stress is the result of an imbalance between the formation of reactive oxygen species (ROS) and the levels of enzymatic and non-enzymatic antioxidants. The assessment of biological redox status is performed by the use of oxidative stress biomarkers. An oxidative stress biomarker is defined as any physical structure or process or chemical compound that can be assessed in a living being (in vivo) or in solid or fluid parts thereof (in vitro), the determination of which is a reproducible and reliable indicator of oxidative stress. The use of oxidative stress biomarkers allows early identification of the risk of developing diseases associated with this process and also opens up possibilities for new treatments. At the end of the last century, interest in oxidative stress biomarkers began to grow, due to evidence of the association between the generation of free radicals and various pathologies. Up to now, a significant number of studies have been carried out to identify and apply different oxidative stress biomarkers in clinical practice. Among the most important oxidative stress biomarkers, it can be mentioned the products of oxidative modifications of lipids, proteins, nucleic acids, and uric acid as well as the measurement of the total antioxidant capacity of fluids in the human body. In this review, we aim to present recent advances and current knowledge on the main biomarkers of oxidative stress, including the discovery of new biomarkers, with emphasis on the various reproductive complications associated with variations in oxidative stress levels.
Collapse
|
43
|
Alshammari GM, Al-Ayed MS, Abdelhalim MA, Al-Harbi LN, Yahya MA. Effects of Antioxidant Combinations on the Renal Toxicity Induced Rats by Gold Nanoparticles. Molecules 2023; 28:molecules28041879. [PMID: 36838869 PMCID: PMC9959587 DOI: 10.3390/molecules28041879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/18/2023] Open
Abstract
This study investigated some possible mechanisms underlying the nephrotoxic effect of gold nanoparticles (AuNPs) in rats and compared the protective effects of selected known antioxidants-namely, melanin, quercetin (QUR), and α-lipoic acid (α-LA). Rats were divided into five treatment groups (eight rats per group): control, AuNPs (50 nm), AuNPs + melanin (100 mg/kg), AuNPs + QUR (200 mg/kg), and AuNPs + α-LA (200 mg/kg). All treatments were administered i.p., daily, for 30 days. AuNPs promoted renal glomerular and tubular damage and impaired kidney function, as indicated by the higher serum levels of creatinine (Cr), urinary flow, and urea and albumin/Cr ratio. They also induced oxidative stress by promoting mitochondrial permeability transition pore (mtPTP) opening, the expression of NOX4, increasing levels of malondialdehyde (MDA), and suppressing glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT). In addition, AuNPs induced renal inflammation and apoptosis, as evidenced by the increase in the total mRNA and the cytoplasmic and nuclear levels of NF-κB, mRNA levels of Bax and caspase-3, and levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Treatment with melanin, QUR, and α-lipoic acid (α-LA) prevented the majority of these renal damage effects of AuNPs and improved kidney structure and function, with QUR being the most powerful. In conclusion, in rats, AuNPs impair kidney function by provoking oxidative stress, inflammation, and apoptosis by suppressing antioxidants, promoting mitochondrial uncoupling, activating NF-κB, and upregulating NOX4. However, QUR remains the most powerful drug to alleviate this toxicity by reversing all of these mechanisms.
Collapse
Affiliation(s)
- Ghedeir M. Alshammari
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
- Correspondence:
| | - Mohammed S. Al-Ayed
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed Anwar Abdelhalim
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Laila Naif Al-Harbi
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Abdo Yahya
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
44
|
Wang Y, Jiang H, Zhang L, Yao P, Wang S, Yang Q. Nanosystems for oxidative stress regulation in the anti-inflammatory therapy of acute kidney injury. Front Bioeng Biotechnol 2023; 11:1120148. [PMID: 36845189 PMCID: PMC9949729 DOI: 10.3389/fbioe.2023.1120148] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Acute kidney injury (AKI) is a clinical syndrome that results from a rapid decline in renal structure or renal functional impairment with the main pathological feature of sublethal and lethal damage to renal tubular cells. However, many potential therapeutic agents cannot achieve the desired therapeutic effect because of their poor pharmacokinetics and short retention time in the kidneys. With the recent emergence and progress of nanotechnology, nanodrugs with unique physicochemical properties could prolong circulation time, enhance efficient targeted delivery, and elevate the accumulation of therapeutics that can cross the glomerular filtration barrier and indicate comprehensive application prospects in the prevention and treatment of AKI. In this review, various types of nanosystems (such as liposomes, polymeric nanosystems, inorganic nanoparticles and cell-derived extracellular vesicles) are designed and applied to improve the pharmacokinetics of drug formation, which could further relieve the burden on the kidneys caused by the final cumulative dose of drugs in conventional treatments. Moreover, the passive or active targeting effect of nanosystems can also reduce the total therapeutic dose and off-target adverse effects on other organs. Nanodelivery systems for treating AKI that alleviate oxidative stress-induced renal cell damage and regulate the inflammatory kidney microenvironment are summarized.
Collapse
Affiliation(s)
- Yue Wang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China,Center of Scientific Research, Chengdu Medical College, Chengdu, Sichuan, China
| | - Hong Jiang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China,Center of Scientific Research, Chengdu Medical College, Chengdu, Sichuan, China
| | - Longyao Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China,Center of Scientific Research, Chengdu Medical College, Chengdu, Sichuan, China
| | - Peng Yao
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Shaoqing Wang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China,*Correspondence: Shaoqing Wang, ; Qian Yang,
| | - Qian Yang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China,Center of Scientific Research, Chengdu Medical College, Chengdu, Sichuan, China,*Correspondence: Shaoqing Wang, ; Qian Yang,
| |
Collapse
|
45
|
Chen E, Wang T, Tu Y, Sun Z, Ding Y, Gu Z, Xiao S. ROS-scavenging biomaterials for periodontitis. J Mater Chem B 2023; 11:482-499. [PMID: 36468674 DOI: 10.1039/d2tb02319a] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Periodontitis is defined as a chronic inflammatory disease in which the continuous activation of oxidative stress surpasses the reactive oxygen species (ROS) scavenging capacity of the endogenous antioxidative defense system. Studies have demonstrated that ROS-scavenging biomaterials should be promising candidates for periodontitis therapy. To benefit the understanding and design of scavenging biomaterials for periodontitis, this review details the relationship between ROS and periodontitis, including direct and indirect damage, the application of ROS-scavenging biomaterials in periodontitis, including organic and inorganic ROS-scavenging biomaterials, and the various dosage forms of fabricated materials currently used for periodontal therapy. Finally, the current situation and further prospects of ROS-scavenging biomaterials in periodontal applications are summarized. Expecting that improved ROS-scavenging biomaterials could be better designed and developed for periodontal and even clinical application.
Collapse
Affiliation(s)
- Enni Chen
- Department of Periodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Tianyou Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Yuan Tu
- Department of Periodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - ZhiYuan Sun
- Department of Periodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Yi Ding
- Department of Periodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Zhipeng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Shimeng Xiao
- Department of Periodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
46
|
Zhang J, Liu H, Yu Q, Zhan Z, Li T, Shu L, Zhang C, Cheng H, Zhang T, Xin H, Wang X. Hair Derived Microneedle Patches for Both Diabetic Foot Ulcer Prevention and Healing. ACS Biomater Sci Eng 2023; 9:363-374. [PMID: 36564012 DOI: 10.1021/acsbiomaterials.2c01333] [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: 12/25/2022]
Abstract
The large amount of reactive oxygen species (ROS) produced by high glucose metabolism in diabetic patients not only induces inflammation but also damages blood vessels, finally resulting in low limb temperature, and the high glucose environment in diabetic patients also makes them susceptible to bacterial infection. Therefore, diabetic foot ulcer (DFU) usually presents as a nonhealing wound. To efficaciously prevent and treat DFU, we proposed a near-infrared (NIR) responsive microneedle (MN) patch hierarchical microparticle (HMP)-ZnO-MN-vascular endothelial growth factor and basic fibroblast growth factor (H-Z-MN-VEGF&bFGF), which could deliver drugs to the limbs painlessly, accurately, and controllably under NIR irradiation. Therein, the hair-derived HMPs exhibited the capacity of scavenging ROS, thereby preventing damage to the blood vessels. Meanwhile, zinc oxide (ZnO) nanoparticles endowed the MN patch with excellent antibacterial activity which could be further enhanced with the photothermal effect of HMPs under NIR irradiation. Moreover, vascular endothelial growth factor and basic fibroblast growth factor could promote the angiogenesis. A series of experiments proved that the MN patch exhibited broad-spectrum antibacterial and anti-inflammatory capacities. In vivo, it obviously increased the temperature of fingertips in diabetic rats as well as promoted collagen deposition and angiogenesis during wound healing. In conclusion, this therapeutic platform provides a promising method for the prevention and treatment of DFU.
Collapse
Affiliation(s)
- Jiao Zhang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi330088, P.R. China
| | - Huijie Liu
- The Affiliated Stomatological Hospital of Nanchang University, The Key Laboratory of Oral Biomedicine, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, Jiangxi330006, P.R. China
| | - Qihang Yu
- College of Chemistry, Nanchang University, Nanchang, Jiangxi330088, P.R. China
| | - Zhenzhen Zhan
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi330088, P.R. China
| | - Tong Li
- College of Pharmacy, Nanchang University, Nanchang, Jiangxi330088, P.R. China
| | - Lingxin Shu
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi330088, P.R. China
| | - Chuxi Zhang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi330088, P.R. China
| | - Haoxin Cheng
- College of Chemistry, Nanchang University, Nanchang, Jiangxi330088, P.R. China
| | - Teng Zhang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi330088, P.R. China
| | - Hongbo Xin
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi330088, P.R. China
| | - Xiaolei Wang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi330088, P.R. China.,College of Chemistry, Nanchang University, Nanchang, Jiangxi330088, P.R. China
| |
Collapse
|
47
|
Liu C, Zou Q, Tang H, Liu J, Zhang S, Fan C, Zhang J, Liu R, Liu Y, Liu R, Zhao Y, Wu Q, Qi Z, Shen Y. Melanin nanoparticles alleviate sepsis-induced myocardial injury by suppressing ferroptosis and inflammation. Bioact Mater 2022; 24:313-321. [PMID: 36632502 PMCID: PMC9813528 DOI: 10.1016/j.bioactmat.2022.12.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Myocardial injury as one of the severe complications leads to the increasing morbidity and mortality in patients with sepsis. Recent studies reported that reactive oxygen species (ROS)-mediated ferroptosis plays a critical role in the development of heart diseases. Therefore, we hypothesized that anti-ferroptosis agent might be a novel potential therapeutic strategy for sepsis-induced cardiac injury. Herein, we demonstrated that a small biocompatible and MRI-visible melanin nanoparticles (MMPP) improves myocardial function by inhibiting ROS-related ferroptosis signaling pathway. In LPS-induced murine sepsis model, after a single dose intravenously injection of MMPP treatment, MMPP markedly alleviated the myocardial injury including cardiac function and heart structure disorder through suppressing iron-accumulation induced ferroptosis. In vitro, MMPP inhibited cardiomyocyte death by attenuating oxidative stress, inflammation and maintaining mitochondrial homeostasis. Collectively, our findings demonstrated that MMPP protected heart against sepsis-induced myocardial injury via inhibiting ferroptosis and inflammation, which might be a novel therapeutic approach in future.
Collapse
Affiliation(s)
- Chang Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China,Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China
| | - Quan Zou
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Huixin Tang
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Jia Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Shiqi Zhang
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Caihong Fan
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Junwei Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Ruiqing Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Yashan Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Ruiyan Liu
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Yan Zhao
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Qiang Wu
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China,Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, 300000, China,Xinjiang Production and Construction Corps Hospital, Xinjiang, 830092, China,Corresponding author. Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China.
| | - Yanna Shen
- School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China,Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China,Corresponding author. School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China.
| |
Collapse
|
48
|
Shi H, Cheng Z. MC1R and melanin-based molecular probes for theranostic of melanoma and beyond. Acta Pharmacol Sin 2022; 43:3034-3044. [PMID: 36008707 PMCID: PMC9712491 DOI: 10.1038/s41401-022-00970-y] [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: 06/19/2022] [Accepted: 07/27/2022] [Indexed: 12/12/2022] Open
Abstract
Malignant melanoma is accounting for most of skin cancer-associated mortality. The incidence of melanoma increased every year worldwide especially in western countries. Treatment efficiency is highly related to the stage of melanoma. Therefore, accurate staging and restaging play a pivotal role in the management of melanoma patients. Though 18F-fluorodeoxyglucose (18F-FDG) positron-emission tomography (PET) has been widely used in imaging of tumor metastases, novel radioactive probes for specific targeted imaging of both primary and metastasized melanoma are still desired. Melanocortin receptor 1 (MC1R) and melanin are two promising biomarkers specifically for melanoma, and numerous research groups including us have been actively developing a plethora of radioactive probes based on targeting of MC1R or melanin for over two decades. In this review, some of the MC1R-targeted tracers and melanin-associated molecular imaging probes developed in our research and others have been briefly summarized, and it provides a quick glance of melanoma-targeted probe design and may contribute to further developing novel molecular probes for cancer theranostics.
Collapse
Affiliation(s)
- Hui Shi
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
49
|
Kwon YS, Zheng M, Zhang AY, Han Z. Melanin-like Nanoparticles as an Alternative to Natural Melanin in Retinal Pigment Epithelium Cells and Their Therapeutic Effects against Age-Related Macular Degeneration. ACS NANO 2022; 16:19412-19422. [PMID: 36269679 DOI: 10.1021/acsnano.2c09087] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Melanin is a natural pigment that is widely distributed in many parts of the human body, such as the skin and retinal pigment epithelium (RPE) in eyes. In contrast to skin melanin, which is being constantly synthesized by the epidermal melanocytes, melanin in the RPE does not regenerate. Melanin is known to function as a potential radical scavenger and photoprotective agent. However, the protective effects of melanin against oxidative stress decline with increasing age. This phenomenon has been correlated with the pathogenesis of age-related macular degeneration (AMD). To increase the potential antioxidant and photoprotective characteristics of melanin, we designed a therapeutic strategy for replenishment of melanin using PEGylated synthetic melanin-like nanoparticles (MNPs) in the RPE for the treatment of AMD. We performed experiments using AMD-like cellular and mouse models and demonstrated that MNPs are biocompatible and selectively target reactive oxygen species (ROS) with powerful antioxidant properties. MNPs can traffic and accumulate in the RPE and are exclusively located in cytosol, but not the nucleus and mitochondria of the cells, for at least 3 months after a single-dose intravitreal injection. Our findings demonstrate that MNPs are able to substitute for natural melanin in the RPE and suggest the potential efficacy of MNPs as a natural radical scavenger against oxidative stress in ROS-related diseases, such as AMD.
Collapse
Affiliation(s)
- Yong-Su Kwon
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina27599, United States
| | - Min Zheng
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina27599, United States
| | - Alice Yang Zhang
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina27599, United States
| | - Zongchao Han
- Department of Ophthalmology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina27599, United States
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina27599, United States
| |
Collapse
|
50
|
Liang S, Tian X, Wang C. Nanozymes in the Treatment of Diseases Caused by Excessive Reactive Oxygen Specie. J Inflamm Res 2022; 15:6307-6328. [PMID: 36411826 PMCID: PMC9675353 DOI: 10.2147/jir.s383239] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/11/2022] [Indexed: 10/29/2023] Open
Abstract
Excessive reactive oxygen species (ROS) may generate deleterious effects on biomolecules, such as DNA damage, protein oxidation and lipid peroxidation, causing cell and tissue damage and eventually leading to the pathogenesis of diseases, such as neurodegenerative diseases, ischemia/reperfusion ((I/R)) injury, and inflammatory diseases. Therefore, the modulation of ROS can be an efficient means to relieve the aforementioned diseases. Several studies have verified that antioxidants such as Mitoquinone (a mitochondrial-targeted coenzyme Q10 derivative) can scavenge ROS and attenuate related diseases. Nanozymes, defined as nanomaterials with intrinsic enzyme-like properties that also possess antioxidant properties, are hence expected to be promising alternatives for the treatment of ROS-related diseases. This review introduces the types of nanozymes with inherent antioxidant activities, elaborates on various strategies (eg, controlling the size or shape of nanozymes, regulating the composition of nanozymes and environmental factors) for modulating their catalytic activities, and summarizes their performances in treating ROS-induced diseases.
Collapse
Affiliation(s)
- Shufeng Liang
- Department of Molecular Biology, Shanxi Province Cancer Hospital/Shanxi Hospital, Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
- Institute of Environmental Sciences, Shanxi University, Taiyuan, People’s Republic of China
| | - Xin Tian
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, People’s Republic of China
| | - Chunyan Wang
- Department of Transfusion, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|