1
|
Li Z, Yu H, Wang Z, Duan H, Li M, Liao J, Yang L. Recent advances in nanotechnology for repairing spinal cord injuries. Biomaterials 2025; 323:123422. [PMID: 40403446 DOI: 10.1016/j.biomaterials.2025.123422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 05/07/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
Spinal cord injury (SCI) remains a formidable clinical challenge with limited therapeutic options. Recent advances in nanotechnology have introduced paradigm-shifting strategies that transcend the limitations of traditional treatments by offering precision, controllability, and multifunctionality in modulating the hostile post-injury microenvironment. This review systematically summarizes nanotechnology-based therapeutic approaches for SCI, including cell-based nanotherapeutics, nanogels/hydrogels, nano-engineered materials, and combinatorial strategies. We emphasize the synergistic design of multifunctional nanoplatforms that integrate neuroprotection, immune modulation, antioxidative capacity, and axonal regeneration within a single system. Special attention is given to microenvironment-responsive smart materials capable of dynamic therapeutic delivery in response to pathological cues. We critically analyze the challenges of clinical translation, such as the need for standardized safety evaluation and personalized therapeutic dosing, and explore emerging solutions including AI-driven nanocarrier design and organoid-based validation. By integrating interdisciplinary innovations, nanotherapies represent an irreplaceable therapeutic paradigm with the potential to achieve spatiotemporal precision and sustained regenerative support for SCI repair.
Collapse
Affiliation(s)
- Zhipeng Li
- The First Affiliated Hospital of China Medical University, Liaoning, 110001, China
| | - Honghao Yu
- Shengjing Hospital of China Medical University, Liaoning, 110004, China
| | - Zhibin Wang
- Shengjing Hospital of China Medical University, Liaoning, 110004, China
| | - Hongmei Duan
- The First Affiliated Hospital of China Medical University, Liaoning, 110001, China
| | - Minglei Li
- Shengjing Hospital of China Medical University, Liaoning, 110004, China
| | - Jun Liao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Lei Yang
- Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| |
Collapse
|
2
|
Yuan Z, Li J, Na Q. Recent advances in biomimetic nanodelivery systems for the treatment of glioblastoma. Colloids Surf B Biointerfaces 2025; 252:114668. [PMID: 40168694 DOI: 10.1016/j.colsurfb.2025.114668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Glioblastoma remain one of the deadliest malignant tumors in the central nervous system, largely due to their aggressiveness, high degree of heterogeneity, and the protective barrier of the blood-brain barrier (BBB). Conventional therapies including surgery, chemotherapy and radiotherapy often fail to improve patient prognosis due to limited drug penetration and non-specific toxicity. We then present recent advances in biomimetic nanodelivery systems, focusing on cell membrane coatings, nanoenzymes, and exosome-based carriers. By mimicking endogenous biological functions, these systems demonstrate improved immune evasion, enhanced BBB traversal, and selective drug release within the tumor microenvironment. Nevertheless, we acknowledge unresolved bottlenecks related to large-scale production, stability, and the intricacies of regulatory compliance. Looking forward, we propose an interdisciplinary roadmap that combines materials engineering, cellular biology, and clinical expertise. Through this collaborative approach, this work aims to optimize biomimetic nanodelivery for glioma therapy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Zhenru Yuan
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Jing Li
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Qi Na
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
3
|
Zhang S, Wang T, Gao T, Liao J, Wang Y, Xu M, Lu C, Liang J, Xu Z, Sun J, Xie Q, Lin Z, Han H. Imaging probes for the detection of brain microenvironment. Colloids Surf B Biointerfaces 2025; 252:114677. [PMID: 40215639 DOI: 10.1016/j.colsurfb.2025.114677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 05/18/2025]
Abstract
The brain microenvironment (BME) is a highly dynamic system that plays a critical role in neural excitation, signal transmission, development, aging, and neurological disorders. BME consists of three key components: neural cells, extracellular spaces, and physical fields, which provide structures and physicochemical properties to synergistically and antagonistically regulate cell behaviors and functions such as nutrient transport, waste metabolism and intercellular communication. Consequently, monitoring the BME is vital to acquire a better understanding of the maintenance of neural homeostasis and the mechanisms underlying neurological diseases. In recent years, researchers have developed a range of imaging probes designed to detect changes in the microenvironment, enabling precise measurements of structural and biophysical parameters in the brain. This advancement aids in the development of improved diagnostic and therapeutic strategies for brain disorders and in the exploration of cutting-edge mechanisms in neuroscience. This review summarizes and highlights recent advances in the probes for sensing and imaging BME. Also, we discuss the design principles, types, applications, challenges, and future directions of probes.
Collapse
Affiliation(s)
- Shiming Zhang
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
| | - Tianyu Wang
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
| | - Tianzi Gao
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
| | - Jun Liao
- Institute of Systems Biomedicine, Department of Pathology, Department of Biophysics School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China
| | - Yang Wang
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
| | - Meng Xu
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
| | - Changyu Lu
- Department of Neurosurgery, Peking University International Hospital, Beijing 102206, PR China
| | - Jianfeng Liang
- Department of Neurosurgery, Peking University International Hospital, Beijing 102206, PR China
| | - Zhengren Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Jianfei Sun
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Qian Xie
- Division of Nephrology, Peking University Third Hospital, Beijing 100096, PR China.
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, Department of Pathology, Department of Biophysics School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, PR China.
| | - Hongbin Han
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China; Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology, Department of Radiology, Peking University Third Hospital, Beijing 100096, PR China.
| |
Collapse
|
4
|
Jiang P, Li J. Recent advances in biomimetic nanodelivery systems for the treatment of depression. Mater Today Bio 2025; 32:101781. [PMID: 40290890 PMCID: PMC12033927 DOI: 10.1016/j.mtbio.2025.101781] [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: 02/23/2025] [Revised: 04/12/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Depression and cognitive disorders remain major challenges in healthcare, with conventional treatments often facing limitations such as slow onset, side effects, and poor drug delivery to the brain. Biomimetic nanodelivery systems, including nanozymes, cell membrane-based systems, and exosomes, have emerged as promising solutions to these issues. These systems leverage natural biological processes to enhance drug targeting, improve bioavailability, and regulate complex biological pathways. Nanoenzymes, with their catalytic properties, offer antioxidant and anti-inflammatory benefits, while cell membranes and exosomes provide efficient targeting and immune evasion. However, challenges remain, including the immaturity of large-scale production techniques, stability concerns, and incomplete understanding of their mechanisms of action. Moreover, the long-term safety, pharmacokinetics, and toxicity of these systems require further investigation. Despite these obstacles, the potential of biomimetic nanodelivery systems to revolutionize depression treatment is significant. Future research should focus on optimizing their preparation, improving drug targeting and release, and ensuring clinical safety. Multidisciplinary collaboration will be essential for advancing these systems from the laboratory to clinical practice, offering new therapeutic avenues for depression and other neurological disorders.
Collapse
Affiliation(s)
- Ping Jiang
- General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Jian Li
- General Hospital of Northern Theater Command, Shenyang, 110016, China
| |
Collapse
|
5
|
Luo Q, Yang J, Yang M, Wang Y, Liu Y, Liu J, Kalvakolanu DV, Cong X, Zhang J, Zhang L, Guo B, Duo Y. Utilization of nanotechnology to surmount the blood-brain barrier in disorders of the central nervous system. Mater Today Bio 2025; 31:101457. [PMID: 39896289 PMCID: PMC11786670 DOI: 10.1016/j.mtbio.2025.101457] [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: 06/28/2024] [Revised: 10/27/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Central nervous system (CNS) diseases are a major cause of disability and death worldwide. Due to the blood-brain barrier (BBB), drug delivery for CNS diseases is extremely challenging. Nano-delivery systems can overcome the limitations of BBB to deliver drugs to the CNS, improve the ability of drugs to target the brain and provide potential therapeutic methods for CNS diseases. At the same time, the choice of different drug delivery methods (bypassing BBB or crossing BBB) can further optimize the therapeutic effect of the nano-drug delivery system. This article reviews the different methods of nano-delivery systems to overcome the way BBB enters the brain. Different kinds of nanoparticles to overcome BBB were discussed in depth.
Collapse
Affiliation(s)
- Qian Luo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Jiaying Yang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Mei Yang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yingtong Wang
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Yiran Liu
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Jixuan Liu
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Dhan V. Kalvakolanu
- Greenebaum NCI Comprehensive Cancer Center, Department of Microbiology and Immunology University of Maryland School Medicine, Baltimore, MD, USA
| | - Xianling Cong
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Jinnan Zhang
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Ling Zhang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Yanhong Duo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
6
|
Pan Y, Li L, Cao N, Liao J, Chen H, Zhang M. Advanced nano delivery system for stem cell therapy for Alzheimer's disease. Biomaterials 2025; 314:122852. [PMID: 39357149 DOI: 10.1016/j.biomaterials.2024.122852] [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: 06/20/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Alzheimer's Disease (AD) represents one of the most significant neurodegenerative challenges of our time, with its increasing prevalence and the lack of curative treatments underscoring an urgent need for innovative therapeutic strategies. Stem cells (SCs) therapy emerges as a promising frontier, offering potential mechanisms for neuroregeneration, neuroprotection, and disease modification in AD. This article provides a comprehensive overview of the current landscape and future directions of stem cell therapy in AD treatment, addressing key aspects such as stem cell migration, differentiation, paracrine effects, and mitochondrial translocation. Despite the promising therapeutic mechanisms of SCs, translating these findings into clinical applications faces substantial hurdles, including production scalability, quality control, ethical concerns, immunogenicity, and regulatory challenges. Furthermore, we delve into emerging trends in stem cell modification and application, highlighting the roles of genetic engineering, biomaterials, and advanced delivery systems. Potential solutions to overcome translational barriers are discussed, emphasizing the importance of interdisciplinary collaboration, regulatory harmonization, and adaptive clinical trial designs. The article concludes with reflections on the future of stem cell therapy in AD, balancing optimism with a pragmatic recognition of the challenges ahead. As we navigate these complexities, the ultimate goal remains to translate stem cell research into safe, effective, and accessible treatments for AD, heralding a new era in the fight against this devastating disease.
Collapse
Affiliation(s)
- Yilong Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| | - Long Li
- Department of Neurosurgery, First Hospital of China Medical University, Liaoning, 110001, China.
| | - Ning Cao
- Army Medical University, Chongqing, 400000, China
| | - Jun Liao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Huiyue Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, 110001, China.
| | - Meng Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| |
Collapse
|
7
|
Wang L, Gu M, Zhang X, Kong T, Liao J, Zhang D, Li J. Recent Advances in Nanoenzymes Based Therapies for Glioblastoma: Overcoming Barriers and Enhancing Targeted Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413367. [PMID: 39854126 PMCID: PMC11905078 DOI: 10.1002/advs.202413367] [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: 10/21/2024] [Revised: 12/14/2024] [Indexed: 01/26/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor originating from glial cells, characterized by high recurrence rates and poor patient prognosis. The heterogeneity and complex biology of GBM, coupled with the protective nature of the blood-brain barrier (BBB), significantly limit the efficacy of traditional therapies. The rapid development of nanoenzyme technology presents a promising therapeutic paradigm for the rational and targeted treatment of GBM. In this review, the underlying mechanisms of GBM pathogenesis are comprehensively discussed, emphasizing the impact of the BBB on treatment strategies. Recent advances in nanoenzyme-based approaches for GBM therapy are explored, highlighting how these nanoenzymes enhance various treatment modalities through their multifunctional capabilities and potential for precise drug delivery. Finally, the challenges and therapeutic prospects of translating nanoenzymes from laboratory research to clinical application, including issues of stability, targeting efficiency, safety, and regulatory hurdles are critically analyzed. By providing a thorough understanding of both the opportunities and obstacles associated with nanoenzyme-based therapies, future research directions are aimed to be informed and contribute to the development of more effective treatments for GBM.
Collapse
Affiliation(s)
- Liyin Wang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Min Gu
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Xiaoli Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | | | - Jun Liao
- Institute of Systems BiomedicineBeijing Key Laboratory of Tumor Systems BiologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Dan Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Jingwu Li
- The First Hospital of China Medical UniversityLiaoning110001China
| |
Collapse
|
8
|
Bi X, Wang Z, He J. Recent advances in biomimetic nanodelivery systems for the treatment of myocardial ischemia reperfusion injury. Colloids Surf B Biointerfaces 2025; 247:114414. [PMID: 39626610 DOI: 10.1016/j.colsurfb.2024.114414] [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: 11/09/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025]
Abstract
Myocardial ischemia/reperfusion injury (MIRI) is a significant challenge in the treatment of myocardial infarction, a leading cause of global mortality due to irreversible cardiac damage. Biomimetic nanodelivery systems offer promising therapeutic strategies to address MIRI. In this review, we comprehensively investigate the underlying pathophysiological mechanisms of MIRI and discuss recent advances in biomimetic nanodelivery systems including cell membrane-coated nanoparticles, exosomes, and nanoenzymes as innovative approaches for MIRI treatment. We emphasize the advantages and potential of biomimetic strategies in enhancing therapeutic efficacy, assess the preclinical effectiveness of these nanodelivery systems, and discuss the challenges associated with translating these approaches into clinical practice. This paper aims to provide new perspectives on biomimetic strategies for MIRI treatment, contributing to the development of effective drug delivery systems.
Collapse
Affiliation(s)
- Xiaojun Bi
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Ze Wang
- Dalian Medical University, Liaoning 116044, China
| | - Jingteng He
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
9
|
He L, Zhang H, Deng J, He Y, Cai Z, He Y. Fluoxetine-induced downregulation of circMap2k1 signaling cascade to improve neurological function after ischemic stroke. Fundam Clin Pharmacol 2025; 39:e13048. [PMID: 39777430 DOI: 10.1111/fcp.13048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 10/14/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Ischemic stroke (IS) is known for its high incidence, disability, and mortality, and there is an urgent need to investigate the pathophysiological mechanisms and develop novel treatment strategies. OBJECTIVES We aimed to investigate the mechanisms of the novel circMap2k1/miR-135b-5p/Pidd1 axis in the treatment of IS progression with fluoxetine. METHODS The middle cerebral artery occlusion (MCAO) model was done in adult male Sprague-Dawley (SD) rats and followed by fluoxetine treatment and the injection of adeno-associated virus (AAV)-sh-ctr and AAV-sh-circMap2k1 into the bilateral hippocampal tissues of rats. Dual-luciferase reporter gene assay was employed to confirm the binding between miR-135b-5p and Pidd1. Enzyme-linked immunosorbent assay was performed to measure the concentrations of the inflammatory factors TNF-α, IL-6, and IL-1β in the plasma. The role of circMap2k1 in cells was tested by overexpression of circMap2k1. Cell viability was assessed using Cell Counting Kit-8 assay, while apoptosis was measured by flow cytometry. RESULTS Knockdown of circMap2k1 enhanced the therapeutic and protective effect of fluoxetine on IS injury in rats. Dual-luciferase reporter gene assay confirmed the targeting of miR-135b-5p to Pidd1. Additionally, fluoxetine deactivated the adsorption of miR-135b-5p by downregulating circMap2k1, and miR-135b-5p further exerted its inhibitory effect on Pidd1 and finally attenuated the inflammatory response caused by microglial polarization after IS. Cell experiments revealed that overexpression of circMap2k1 repressed cell viability and promoted cell apoptosis. CONCLUSIONS Fluoxetine downregulated of circMap2k1 was associated ameliorate neurological injury and inflammatory responses induced by microglial polarization after IS. The manuscript is available as a preprint at this link: doi.org/10.21203/rs.3.rs-3209057/v1.
Collapse
Affiliation(s)
- Langtao He
- Department of Geriatrics, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Hui Zhang
- Department of Neurology, The Second Clinical Medical College of Jinan University, Shenzhen, China
- Department of Neurology, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
- Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
| | - Jian Deng
- Department of Neurology, The Second Clinical Medical College of Jinan University, Shenzhen, China
- Department of Neurology, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
- Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
| | - Yibo He
- Department of Neurology, The Second Clinical Medical College of Jinan University, Shenzhen, China
- Department of Neurology, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
- Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
| | - Zhili Cai
- Department of Neurology, The Second Clinical Medical College of Jinan University, Shenzhen, China
- Department of Neurology, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
- Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
| | - Yitao He
- Department of Neurology, The Second Clinical Medical College of Jinan University, Shenzhen, China
- Department of Neurology, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
- Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
| |
Collapse
|
10
|
Ruan L, Zheng M, Xia X, Pang C, Wang Y, Fan Z, Yang J, Qing Q, Lin H, Tao Y, Wang J, Wang L. Exploring the Constituents and Mechanisms of Polygonum multiflorum Thunb. in Mitigating Ischemic Stroke: A Network Pharmacology and Molecular Docking Study. Comb Chem High Throughput Screen 2025; 28:781-797. [PMID: 38623977 DOI: 10.2174/0113862073285988240229081558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 04/17/2024]
Abstract
BACKGOUND Polygonum multiflorum Thunb. (PMT) has shown promise in exerting cerebrovascular protective effects, and its potential for treating ischemic stroke (IS) has garnered attention. However, the lack of clarity regarding its chemical constituents and mechanisms has significantly hindered its clinical application. METHODS In this study, we employed network pharmacology and molecular docking techniques for the first time to elucidate the potential compounds and targets of PMT in treating IS. The databases CTD, DrugBank, DisGeNET, GeneCards, OMIM, TTD, PGKB, NCBI, TCMIP, CNKI, PubMed, ZINC, STITCH, BATMAN, ETCM and Swiss provided information on targets related to IS and components of PMT, along with their associated targets. We constructed "compoundtarget" and protein-protein interaction (PPI) networks sourced from the STRING database using the Cytoscape software. Gene Ontology (GO) enrichment analysis and KEGG pathway analysis were conducted using the DAVID database. Molecular docking between core targets and active compounds was conducted using Autodock4 software. Experiments were performed in an oxygen- glucose deprivation and reperfusion (OGD/R) model to validate the anti-IS activity of compounds isolated from PMT preliminarily. Network pharmacological analysis revealed 16 core compounds, including resveratrol, polydatin, TSG, ω-hydroxyemodin, emodin anthrone, tricin, moupinamide, and others, along with 11 high-degree targets, such as PTGS1, PTGS2, ADORA1, ADORA2, CA1, EGFR, ESR1, ESR2, SRC, MMP3 and MMP9. RESULTS GO and KEGG enrichment analyses revealed the involvement of HIF-1, Akt signaling pathway and energy metabolism-related signaling pathways. Molecular docking results emphasized eight key compounds and confirmed their interactions with corresponding targets. In vitro OGD/R model experiments identified TSG and tricin as the primary active substances within PMT for its anti-stroke activity. CONCLUSION This study contributes new insights into the potential development of PMT for stroke prevention and treatment.
Collapse
Affiliation(s)
- Lingyu Ruan
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Mengyun Zheng
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Xinru Xia
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Chaofan Pang
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Yating Wang
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Zhiwei Fan
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Jingtian Yang
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Qing Qing
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Hongyan Lin
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Yuheng Tao
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Junsong Wang
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Liqun Wang
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| |
Collapse
|
11
|
Belgamwar A, Sharma R, Mali Y, Agrawal YO, Nakhate KT. Nano revolutions in ischemic stroke: A critical analysis of current options and the potential of nanomedicines in diagnosis and therapeutics. Neuroscience 2024; 562:90-105. [PMID: 39433081 DOI: 10.1016/j.neuroscience.2024.10.022] [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: 09/13/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
A stroke, also known as cerebrovascular accident, is a medical emergency that occurs when the blood supply to the brain is interrupted. This disruption can happen in two main ways: through a hemorrhagic stroke, where a blood vessel in the brain bursts, or through an ischemic stroke, where a blood clot blocks an artery. Both types of stroke cause damage to brain cells, leading to a range of health complications. Globally, stroke ranks as the second leading cause of death and disability.This review provides an overview of stroke, focusing on its early detection, current treatment options, and emerging therapies. We discuss the complex mechanisms that contribute to stroke development, including the roles of cells, biomolecules, and blood vessels. Additionally, the review explores recent advances in the use of nanoparticles to enhance the efficacy of the pharmacotherapy of stroke, particularly ischemic stroke. Ongoing clinical trials in stroke management are also highlighted. Timely diagnosis and prompt intervention are critical for improving patient outcomes.We aim to increase awareness and understanding of stroke among researchers and healthcare professionals, ultimately improving patient care.
Collapse
Affiliation(s)
- Aarti Belgamwar
- Department of Pharmaceutics, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra 424001, India
| | - Rarchita Sharma
- Department of Pharmaceutics, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra 424001, India
| | - Yogesh Mali
- Department of Pharmaceutics, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra 424001, India
| | - Yogeeta O Agrawal
- Department of Pharmaceutics, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra 424001, India.
| | - Kartik T Nakhate
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra 424001, India
| |
Collapse
|
12
|
Bi X, Cao N, He J. Recent advances in nanoenzymes for Alzheimer's disease treatment. Colloids Surf B Biointerfaces 2024; 244:114139. [PMID: 39121571 DOI: 10.1016/j.colsurfb.2024.114139] [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: 06/04/2024] [Revised: 07/14/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Alzheimer's disease (AD) remains one of the most challenging neurodegenerative disorders to treat, with oxidative stress playing a significant role in its pathology. Recent advancements in nanoenzymes technology offer a promising approach to mitigate this oxidative damage. Nanoenzymes, with their unique enzyme-mimicking activities, effectively scavenge reactive oxygen species and reduce oxidative stress, thereby providing neuroprotective effects. This review delves into the underlying mechanisms of AD, focusing on oxidative stress and its impact on disease progression. We explore the latest developments in nanoenzymes applications for AD treatment, highlighting their multifunctional capabilities and potential for targeted delivery to amyloid-beta plaques. Despite the exciting prospects, the clinical translation of nanoenzymes faces several challenges, including difficulties in brain targeting, consistent quality production, and ensuring safety and biocompatibility. We discuss these limitations in detail, emphasizing the need for rigorous evaluation and standardized protocols. This paper aims to provide a comprehensive overview of the current state of nanoenzymes research in AD, shedding light on both the opportunities and obstacles in the path towards effective clinical applications.
Collapse
Affiliation(s)
- Xiaojun Bi
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Ning Cao
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing 400000, China
| | - Jingteng He
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
13
|
Hou D, Hu Y, Yun T, Li H, Yang G, Yu D. The deubiquitinase OTUD3 stabilizes IRP2 expression to reduce hippocampal neuron ferroptosis via the p53/PTGS2 pathway to ameliorate cerebral ischemia-reperfusion injury. Eur J Med Res 2024; 29:498. [PMID: 39415292 PMCID: PMC11484114 DOI: 10.1186/s40001-024-02095-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Ischemic stroke (IS) is known for its high morbidity, disability and mortality rates, and studies designed to explore its pathophysiological mechanisms and identify novel therapeutic strategies are urgently needed. We aimed to probe the effects of the deubiquitinase OTUD3-IRP2-p53/PTGS2 pathway on cerebral ischemia‒reperfusion (I/R) injury and hippocampal neuron ferroptosis. METHODS A cerebral I/R mouse model was established. Furthermore, lentiviral vectors that overexpressed OTUD3 and knocked down IRP2 were constructed, and a series of assays were performed to probe the OTUD3/IRP2/p53/PTGS2 mechanism. An oxygen‒glucose deprivation and reoxygenation (OGD/R) model of mouse hippocampal neurons was constructed. Then, OTUD3 and IRP2 were knocked down and overexpressed, and p53 was overexpressed to explore the mechanism of the OTUD3/IRP2/p53/PTGS2 pathway. RESULTS OTUD3 and IRP2 were expressed at low levels in cerebral I/R models. OTUD3 promoted IRP2 expression to protect damaged hippocampal neurons. Moreover, IRP2 affected ferroptosis in hippocampal neurons. In addition, IRP2 inhibited p53. After IRP2 and p53 were overexpressed, IRP2 regulated the p53/PTGS2 pathway and affected ferroptosis in hippocampal neurons. In vivo, after overexpressing OTUD3 and knocking down IRP2, we found that overexpression of OTUD3 promoted IRP2 expression to reduce ferroptosis in hippocampal neurons and improve cerebral I/R injury via the inhibition of the p53/PTGS2 pathway. CONCLUSIONS The deubiquitinase OTUD3 stabilized IRP2 expression to reduce hippocampal neuron ferroptosis via the p53/PTGS2 pathway to subsequently ameliorate cerebral I/R injury.
Collapse
Affiliation(s)
- Dan Hou
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, Hainan, China
| | - Yujie Hu
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, Hainan, China
| | - Tian Yun
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, Hainan, China
| | - Hongxin Li
- School of Statistics Major, Beijing Forestry University, Beijing, 100091, China
| | - Guoshuai Yang
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, Hainan, China.
| | - Dan Yu
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, Hainan, China.
| |
Collapse
|
14
|
Wang C, Xiao Z, Fan J, Zhang C, Wang T, Qiu Z, Ye F, Chen M, Li Y. Nanocarriers Loaded with Danshensu for Treating Ischemic Stroke by Reducing Oxidative Stress and Glial Overactivation. ACS OMEGA 2024; 9:35686-35694. [PMID: 39184494 PMCID: PMC11339989 DOI: 10.1021/acsomega.4c03991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024]
Abstract
Ischemic stroke is a complex health condition that can cause ischemia and necrosis of brain tissue. Subsequently, the excessive activation of glial cells can result in various inflammatory and oxidative stress reactions that exacerbate ischemic brain injury. In this paper, we propose the targeted self-assembly of a three-dimensional nanoparticle network containing Danshensu to rescue ischemic penumbra by reducing oxidative stress and glial overactivation. The network comprises nanoparticles composed of chitosan, thiol ketone, and carboxymethyl-β-cyclodextrin as the core wrapped by the Pro-His-Ser-Arg-Asn (PHSRN) peptide sequence as the outer layer and loaded with Danshensu. The PHSRN-peptide-modified nanoparticles bind to integrin α5β1 overexpressed on the damaged blood-brain barrier and accumulate in the damaged brain in a rat model of ischemia/reperfusion. When stimulated by reactive oxygen species, thiol ketone bonded to the nanoparticles was hydrolyzed, facilitating responsive drug release while consuming the reactive oxygen species. Subsequently, the released Danshensu scavenged the reactive oxygen species to prevent oxidative stress and inhibited the activation of astrocytes, thereby suppressing proinflammatory cytokine secretion, improving the inflammatory brain microenvironment and reducing neuronal apoptosis.
Collapse
Affiliation(s)
- Cuihong Wang
- Department
of Pharmacy, Shanghai University of Medicine
and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Zhicheng Xiao
- 411
Hospital of Shanghai University, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Jinhui Fan
- 411
Hospital of Shanghai University, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Chuan Zhang
- 411
Hospital of Shanghai University, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Tingfang Wang
- 411
Hospital of Shanghai University, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Zheng Qiu
- Shenzhen
Medicines and Health Products IMP. & EXP. Co., Ltd., Guangdong 518000, China
| | - Fei Ye
- 411
Hospital of Shanghai University, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Min Chen
- Department
of Pharmacy, Shanghai University of Medicine
and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Yi Li
- 411
Hospital of Shanghai University, School of Medicine, Shanghai University, Shanghai 200444, China
| |
Collapse
|
15
|
Liu W, Liu L, Li H, Xie Y, Bai J, Guan J, Qi H, Sun J. Targeted pathophysiological treatment of ischemic stroke using nanoparticle-based drug delivery system. J Nanobiotechnology 2024; 22:499. [PMID: 39164747 PMCID: PMC11337765 DOI: 10.1186/s12951-024-02772-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
Ischemic stroke poses significant challenges in terms of mortality and disability rates globally. A key obstacle to the successful treatment of ischemic stroke lies in the limited efficacy of administering therapeutic agents. Leveraging the unique properties of nanoparticles for brain targeting and crossing the blood-brain barrier, researchers have engineered diverse nanoparticle-based drug delivery systems to improve the therapeutic outcomes of ischemic stroke. This review provides a concise overview of the pathophysiological mechanisms implicated in ischemic stroke, encompassing oxidative stress, glutamate excitotoxicity, neuroinflammation, and cell death, to elucidate potential targets for nanoparticle-based drug delivery systems. Furthermore, the review outlines the classification of nanoparticle-based drug delivery systems according to these distinct physiological processes. This categorization aids in identifying the attributes and commonalities of nanoparticles that target specific pathophysiological pathways in ischemic stroke, thereby facilitating the advancement of nanomedicine development. The review discusses the potential benefits and existing challenges associated with employing nanoparticles in the treatment of ischemic stroke, offering new perspectives on designing efficacious nanoparticles to enhance ischemic stroke treatment outcomes.
Collapse
Affiliation(s)
- Wei Liu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Lubin Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Hong Li
- Clinical Laboratory, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, 266033, China
| | - Yutong Xie
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Ju Bai
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Jialiang Guan
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Jinping Sun
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
16
|
Liao J, Gong L, Xu Q, Wang J, Yang Y, Zhang S, Dong J, Lin K, Liang Z, Sun Y, Mu Y, Chen Z, Lu Y, Zhang Q, Lin Z. Revolutionizing Neurocare: Biomimetic Nanodelivery Via Cell Membranes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402445. [PMID: 38583077 DOI: 10.1002/adma.202402445] [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: 02/16/2024] [Revised: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Brain disorders represent a significant challenge in medical science due to the formidable blood-brain barrier (BBB), which severely limits the penetration of conventional therapeutics, hindering effective treatment strategies. This review delves into the innovative realm of biomimetic nanodelivery systems, including stem cell-derived nanoghosts, tumor cell membrane-coated nanoparticles, and erythrocyte membrane-based carriers, highlighting their potential to circumvent the BBB's restrictions. By mimicking native cell properties, these nanocarriers emerge as a promising solution for enhancing drug delivery to the brain, offering a strategic advantage in overcoming the barrier's selective permeability. The unique benefits of leveraging cell membranes from various sources is evaluated and advanced technologies for fabricating cell membrane-encapsulated nanoparticles capable of masquerading as endogenous cells are examined. This enables the targeted delivery of a broad spectrum of therapeutic agents, ranging from small molecule drugs to proteins, thereby providing an innovative approach to neurocare. Further, the review contrasts the capabilities and limitations of these biomimetic nanocarriers with traditional delivery methods, underlining their potential to enable targeted, sustained, and minimally invasive treatment modalities. This review is concluded with a perspective on the clinical translation of these biomimetic systems, underscoring their transformative impact on the therapeutic landscape for intractable brain diseases.
Collapse
Affiliation(s)
- Jun Liao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Lidong Gong
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Qingqiang Xu
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Jingya Wang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yuanyuan Yang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Shiming Zhang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Junwei Dong
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Kerui Lin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Zichao Liang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yuhan Sun
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Yongxu Mu
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014040, China
| | - Zhengju Chen
- Pooling Medical Research Institutes of 100Biotech, Beijing, 100006, China
| | - Ying Lu
- Department of Pharmaceutics, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Qiang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
17
|
Wang G, Li Z, Wang G, Sun Q, Lin P, Wang Q, Zhang H, Wang Y, Zhang T, Cui F, Zhong Z. Advances in Engineered Nanoparticles for the Treatment of Ischemic Stroke by Enhancing Angiogenesis. Int J Nanomedicine 2024; 19:4377-4409. [PMID: 38774029 PMCID: PMC11108071 DOI: 10.2147/ijn.s463333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
Angiogenesis, or the formation of new blood vessels, is a natural defensive mechanism that aids in the restoration of oxygen and nutrition delivery to injured brain tissue after an ischemic stroke. Angiogenesis, by increasing vessel development, may maintain brain perfusion, enabling neuronal survival, brain plasticity, and neurologic recovery. Induction of angiogenesis and the formation of new vessels aid in neurorepair processes such as neurogenesis and synaptogenesis. Advanced nano drug delivery systems hold promise for treatment stroke by facilitating efficient transportation across the the blood-brain barrier and maintaining optimal drug concentrations. Nanoparticle has recently been shown to greatly boost angiogenesis and decrease vascular permeability, as well as improve neuroplasticity and neurological recovery after ischemic stroke. We describe current breakthroughs in the development of nanoparticle-based treatments for better angiogenesis therapy for ischemic stroke employing polymeric nanoparticles, liposomes, inorganic nanoparticles, and biomimetic nanoparticles in this study. We outline new nanoparticles in detail, review the hurdles and strategies for conveying nanoparticle to lesions, and demonstrate the most recent advances in nanoparticle in angiogenesis for stroke treatment.
Collapse
Affiliation(s)
- Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Gongchen Wang
- Department of Vascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Qixu Sun
- Department of Gastroenterology, Penglai People’s Hospital, Yantai, Shandong, 265600, People’s Republic of China
| | - Peng Lin
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Qian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Huishu Zhang
- Teaching Center of Biotechnology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Tongshuai Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Feiyun Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhaohua Zhong
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
18
|
Li Y, Liao J, Xiong L, Xiao Z, Ye F, Wang Y, Chen T, Huang L, Chen M, Chen ZS, Wang T, Zhang C, Lu Y. Stepwise targeted strategies for improving neurological function by inhibiting oxidative stress levels and inflammation following ischemic stroke. J Control Release 2024; 368:607-622. [PMID: 38423472 DOI: 10.1016/j.jconrel.2024.02.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/01/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
Ischemia-reperfusion injury is caused by excessive production of reactive oxygen species (ROS) and inflammation accompanied by ischemic injury symptoms and blood-brain barrier (BBB) dysfunction. This causes neuronal damage, for which no effective treatments or drugs exist. Herein, we provided a stepwise targeted drug delivery strategy and successfully prepared multifunctional ORD@SHp@ANG nanoparticles (NPs) that consist of a stroke homing peptide (DSPE-PEG2000-SHp), BBB-targeting peptide (DSPE-PEG2000-ANG), and ROS-responsive Danshensu (salvianic acid A) chain self-assembly. ORD@SHp@ANG NPs effectively crossed the BBB by ANG peptide and selectively targeted the ischemic brain sites using stroke-homing peptide. The results showed that ORD@SHp@ANG NPs can effective at scavenging ROS, and protect SH-SY5Y cells from oxidative damage in vitro. Furthermore, ORD@SHp@ANG NPs showed excellent biocompatibility. These NPs recognized brain endothelial cells and crossed the BBB, regulated the transformation of microglia into the anti-inflammatory phenotype, and inhibited the production of inflammatory factors in a rat ischemia-reperfusion model, thereby reducing cerebral infarction, neuronal apoptosis and preserving BBB integrity. Sequencing revealed that ORD@SHp@ANG NPs promote cell proliferation, activate immune responses, suppress inflammatory responses, and ameliorate ischemic stroke. In conclusion, this study reports a simple and promising drug delivery strategy for managing ischemic stroke.
Collapse
Affiliation(s)
- Yi Li
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, or Materials Science and Engineering, Shanghai University, Shanghai 200444, China; Department of Pharmacy, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Jun Liao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, or Materials Science and Engineering, Shanghai University, Shanghai 200444, China; Department of Pharmaceutical Science, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Liyan Xiong
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, or Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Zhicheng Xiao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, or Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Fei Ye
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, or Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Yun Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, or Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Ting Chen
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, or Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Linzhang Huang
- Institute of Metabolic and Integrative Biology, Fudan University, Shanghai 201399, China
| | - Min Chen
- Department of Pharmacy, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York 11439, USA.
| | - Tingfang Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, or Materials Science and Engineering, Shanghai University, Shanghai 200444, China.
| | - Chuan Zhang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, or Materials Science and Engineering, Shanghai University, Shanghai 200444, China.
| | - Ying Lu
- Department of Pharmaceutical Science, School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
19
|
Fan L, Lin X, Hong L, Li L, Lin R, Ren T, Tian J, Chen M. Simultaneous antioxidant and neuroprotective effects of two-dimensional (2D) MXene-loaded isoquercetin for ischemic stroke treatment. J Mater Chem B 2024; 12:2795-2806. [PMID: 38385522 DOI: 10.1039/d3tb01973j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Oxidative stress and reactive oxygen species drive ischemic stroke and its related complications. New antioxidant medications are therefore crucial for treating ischemic stroke. We developed Ti2C@BSA-ISO nanocomposites loaded with the hydrophobic drug isoquercetin (ISO) encapsulated in BSA on Ti2C nano-enzymes as a novel therapeutic nanomedicine for the treatment of ischemic stroke targeting reactive oxygen species (ROS). TEM visually proved the successful preparation of Ti2C@BSA-ISO, and the FTIR, XPS, zeta potential and DLS together demonstrated the acquisition of Ti2C@BSA-ISO. In addition, the enzyme-mimicking activity of Ti2C was evaluated and the antioxidant capacity of Ti2C@BSA-ISO was verified. Ti2C@BSA-ISO was able to reverse the decrease in cellular activity caused by ROS. Experiments in vivo showed that Ti2C@BSA-ISO could promote neuroprotection and scavenging of ROS in the hippocampal CA1 area and cerebral cortex of rats, thereby inhibiting cellular death and alleviating ischaemic stroke. Specifically, Ti2C@BSA-ISO alleviated ischemic stroke by inhibiting NLRP3/caspase-1/GSDMD pathway-mediated pyroptosis. Our study demonstrates the effectiveness of nanomedicines that can be directly used as drugs for the treatment of ischemic stroke in synergy with other drugs, which greatly expands the application of nanomaterials in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Limin Fan
- Department of Emergency, The First Affiliated Hospital of Hainan Medical University, No. 31, Longhua Road, Longhua District, Haikou City, Hainan Province, 570102, P. R. China.
- School of Medicine, Tongji University, Shanghai 200092, P. R. China
| | - Xinhua Lin
- Department of Emergency, The First Affiliated Hospital of Hainan Medical University, No. 31, Longhua Road, Longhua District, Haikou City, Hainan Province, 570102, P. R. China.
| | - Limin Hong
- Department of Emergency, The First Affiliated Hospital of Hainan Medical University, No. 31, Longhua Road, Longhua District, Haikou City, Hainan Province, 570102, P. R. China.
| | - Lehui Li
- Department of Emergency, The First Affiliated Hospital of Hainan Medical University, No. 31, Longhua Road, Longhua District, Haikou City, Hainan Province, 570102, P. R. China.
| | - Run Lin
- Department of Emergency, The First Affiliated Hospital of Hainan Medical University, No. 31, Longhua Road, Longhua District, Haikou City, Hainan Province, 570102, P. R. China.
| | - Tianbin Ren
- School of Medicine, Tongji University, Shanghai 200092, P. R. China
| | - Jia Tian
- Intensive Medical Unit, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19 Xiuhua Road, Haikou, China.
| | - Miao Chen
- Department of Emergency, The First Affiliated Hospital of Hainan Medical University, No. 31, Longhua Road, Longhua District, Haikou City, Hainan Province, 570102, P. R. China.
| |
Collapse
|
20
|
Xu ZL, Li CJ, Qian X, Duan H, Zhou J, Zhang QQ, Dong X, Zhao L. A validated LC-MS/MS method for determination of six Anti-SARS-CoV-2 drugs in plasma and its application for a pharmacokinetic study in rats. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1235:124038. [PMID: 38341953 DOI: 10.1016/j.jchromb.2024.124038] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/13/2024]
Abstract
Antiviral treatment for COVID-19 is considered an effective tool in reducing the rate of severe cases and deaths. As of June 2023, a total of six small molecule antiviral drugs have been conditionally approved for marketing by the National Medical Products Administration (NMPA) within China. In this study, a method of HPLC-MS/MS was established and validated for the determination of six small molecule antiviral drugs in plasma using Lamivudine as an internal standard. The chromatographic separation was performed using gradient elution with an ACE 3 C18-PFP column (3.0 mm × 150 mm, 3 μm), and the mobile phase consisted of deionized water and acetonitrile/water (90:10, v/v), both with 10 mmol/L of ammonium acetate and 0.1 % ammonium hydroxide added. Quantitative analysis of the six small molecule drugs was carried out through selective reaction monitoring based on the positive ion spray ionization mode. The method exhibited excellent precision, accuracy, recovery, and linearity, and it was used to determine the pharmacokinetic characteristics in rats. Our work not only established a bioanalytical method for six small molecule antiviral drugs but also provided scientific references for clinical pharmacokinetic studies.
Collapse
Affiliation(s)
- Zong-Liang Xu
- School of Medicine, Shanghai University, Shanghai 200444, China; Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai 201908, China
| | - Cheng-Jian Li
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai 201908, China
| | - Xian Qian
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai 201908, China
| | - Hu Duan
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai 201908, China
| | - Jin Zhou
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai 201908, China
| | - Qian-Qian Zhang
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai 201908, China
| | - Xin Dong
- School of Medicine, Shanghai University, Shanghai 200444, China; Suzhou lnnovation Center of Shanghai University, Suzhou, Jiangsu, China.
| | - Liang Zhao
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai 201908, China.
| |
Collapse
|
21
|
Liao J, Li Y, Fan L, Sun Y, Gu Z, Xu QQ, Wang Y, Xiong L, Xiao K, Chen ZS, Ma Z, Zhang C, Wang T, Lu Y. Bioactive Ceria Nanoenzymes Target Mitochondria in Reperfusion Injury to Treat Ischemic Stroke. ACS NANO 2024. [PMID: 38266247 DOI: 10.1021/acsnano.3c10982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Overproduction of reactive oxygen species by damaged mitochondria after ischemia is a key factor in the subsequent cascade of damage. Delivery of therapeutic agents to the mitochondria of damaged neurons in the brain is a potentially promising targeted therapeutic strategy for the treatment of ischemic stroke. In this study, we developed a ceria nanoenzymes synergistic drug-carrying nanosystem targeting mitochondria to address multiple factors of ischemic stroke. Each component of this nanosystem works individually as well as synergistically, resulting in a comprehensive therapy. Alleviation of oxidative stress and modulation of the mitochondrial microenvironment into a favorable state for ischemic tolerance are combined to restore the ischemic microenvironment by bridging mitochondrial and multiple injuries. This work also revealed the detailed mechanisms by which the proposed nanodelivery system protects the brain, which represents a paradigm shift in ischemic stroke treatment.
Collapse
Affiliation(s)
- Jun Liao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Pharmaceutical Sciences, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Yi Li
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Li Fan
- Department of Pharmaceutical Sciences, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Yuhan Sun
- Department of Pharmaceutical Sciences, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Zhengyan Gu
- Department of Pharmaceutical Sciences, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Qing-Qiang Xu
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Yun Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Liyan Xiong
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Kai Xiao
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York 11439, United States
| | - Zhiwei Ma
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, 201210, China
| | - Chuan Zhang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Tingfang Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Ying Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
22
|
Hao C, Sha M, Ye Y, Wang C. Cell Membrane-Derived Nanovehicles for Targeted Therapy of Ischemic Stroke: From Construction to Application. Pharmaceutics 2023; 16:6. [PMID: 38276484 PMCID: PMC10819970 DOI: 10.3390/pharmaceutics16010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 01/27/2024] Open
Abstract
Ischemic stroke (IS) is a prevalent form of stroke and a leading cause of mortality and disability. Recently, cell membrane-derived nanovehicles (CMNVs) derived from erythrocytes, thrombocytes, neutrophils, macrophages, neural stem cells, and cancer cells have shown great promise as drug delivery systems for IS treatment. By precisely controlling drug release rates and targeting specific sites in the brain, CMNVs enable the reduction in drug dosage and minimization of side effects, thus significantly enhancing therapeutic strategies and approaches for IS. While there are some reviews regarding the applications of CMNVs in the treatment of IS, there has been limited attention given to important aspects such as carrier construction, structural design, and functional modification. Therefore, this review aims to address these key issues in CMNVs preparation, structural composition, modification, and other relevant aspects, with a specific focus on targeted therapy for IS. Finally, the challenges and prospects in this field are discussed.
Collapse
Affiliation(s)
- Cui Hao
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (H.C.); (S.M.); (Y.Y.)
| | - Ma Sha
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (H.C.); (S.M.); (Y.Y.)
| | - Yang Ye
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (H.C.); (S.M.); (Y.Y.)
- Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan Province, Kunming 650500, China
| | - Chengxiao Wang
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (H.C.); (S.M.); (Y.Y.)
- Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan Province, Kunming 650500, China
| |
Collapse
|
23
|
Tang L, Fu C, Zhang A, Li X, Cao Y, Feng J, Liu H, Dong H, Wang W. Harnessing nanobiotechnology for cerebral ischemic stroke management. Biomater Sci 2023; 11:791-812. [PMID: 36545758 DOI: 10.1039/d2bm01790c] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cerebral ischemic stroke remains one of the most serious neurological disorders that pose threats to human health, causing a large amount of long-term disability or even death throughout the world. Based on its physiologic and pathological features, there are limited available therapeutic options for effective ischemic stroke management. Encouragingly, a rapid advancement of nanobiotechnology is bringing new insights into exploring more alternative strategies against cerebral ischemic stroke, which can cleverly overcome the limitations related to conventional treatment methods. Therefore, this review focuses on the recent achievements of nanobiotechnology for ischemic stroke management, which emphasizes diverse targeted delivery strategies using various nanoplatforms including liposomes, micelles, polymeric nanoparticles, nanogels, inorganic nanomaterials, and cell-derived nano-vectors based on the pathophysiological features of ischemic stroke. Moreover, different therapeutic approaches against ischemic stroke such as neuroprotection, anti-inflammation, thrombolysis, increased blood-brain barrier penetration and reactive oxygen species scavenging are highlighted. Meanwhile, this review discusses how these versatile nanoplatforms were designed to assist in the treatment of ischemic stroke. Based on this, challenges, opportunities, and future perspectives using nanobiotechnology through rational design for effective ischemic stroke management are revealed.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China. .,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Cong Fu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China. .,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Aining Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China. .,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Xiyue Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China. .,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Yuqi Cao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China. .,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Jingwen Feng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China. .,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Hening Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China. .,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Haijuan Dong
- The Public Laboratory Platform, China Pharmaceutical University, 210009 Nanjing, China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China. .,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| |
Collapse
|
24
|
Yu Q, Jian Z, Yang D, Zhu T. Perspective insights into hydrogels and nanomaterials for ischemic stroke. Front Cell Neurosci 2023; 16:1058753. [PMID: 36761147 PMCID: PMC9902513 DOI: 10.3389/fncel.2022.1058753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/30/2022] [Indexed: 01/26/2023] Open
Abstract
Ischemic stroke (IS) is a neurological disorder prevalent worldwide with a high disability and mortality rate. In the clinic setting, tissue plasminogen activator (tPA) and thrombectomy could restore blood flow of the occlusion region and improve the outcomes of IS patients; however, these therapies are restricted by a narrow time window. Although several preclinical trials have revealed the molecular and cellular mechanisms underlying infarct lesions, the translatability of most findings is unsatisfactory, which contributes to the emergence of new biomaterials, such as hydrogels and nanomaterials, for the treatment of IS. Biomaterials function as structural scaffolds or are combined with other compounds to release therapeutic drugs. Biomaterial-mediated drug delivery approaches could optimize the therapeutic effects based on their brain-targeting property, biocompatibility, and functionality. This review summarizes the advances in biomaterials in the last several years, aiming to discuss the therapeutic potential of new biomaterials from the bench to bedside. The promising prospects of new biomaterials indicate the possibility of an organic combination between materialogy and medicine, which is a novel field under exploration.
Collapse
Affiliation(s)
- Qingbo Yu
- Laboratory of Anesthesia & Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China,Department of Anesthesiology, North Sichuan Medical College, Nanchong, China
| | - Zhang Jian
- Sichuan Provincial Maternity and Child Health Care Hospital, Women’s and Children’s Hospital Affiliated of Chengdu Medical College, Chengdu, China
| | - Dan Yang
- Department of Anesthesiology, North Sichuan Medical College, Nanchong, China
| | - Tao Zhu
- Laboratory of Anesthesia & Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China,*Correspondence: Tao Zhu,
| |
Collapse
|