1
|
Zhao Y, Gai C, Yu S, Song Y, Gu B, Luo Q, Wang X, Hu Q, Liu W, Liu D, Wang Z. Liposomes-Loaded miR-9-5p Alleviated Hypoxia-Ischemia-Induced Mitochondrial Oxidative Stress by Targeting ZBTB20 to Inhibiting Nrf2/Keap1 Interaction in Neonatal Mice. Antioxid Redox Signal 2025; 42:512-528. [PMID: 39869050 DOI: 10.1089/ars.2024.0640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Aims: Hypoxia ischemia (HI) is a leading cause of cerebral palsy and long-term neurological sequelae in infants. Given that mitochondrial dysfunction in neurons contributes to HI brain damage, this study aimed to investigate the regulatory role of miR-9-5p in mitochondrial function following HI injury. Results: Overexpression of miR-9-5p in HI mice or H2O2-exposed PC12 cells suppressed neuronal injury, associated with increased mitochondrial copy number, normalizing mitochondrial membrane potential, improved nuclear factor-erythroid factor 2-related factor 2 (Nrf2) activation, and downregulation of Keap1. This was mediated, in part, through the ability of this miR-9-5p to bind and regulate the transcriptional activity of zinc finger and BTB domain-containing protein 20 (ZBTB20). Further study suggested that the knockdown of ZBTB20 exerts neuroprotection by inhibiting Nrf2/Keap1 interaction to promote the translocation of Nrf2 from the cytoplasm to the nucleus and the consequent expression of antioxidant proteins. Notably, the protective effects of miR-9-5p overexpression against HI-induced mitochondrial damage were reversed by the Nrf2 inhibitor ML385. Finally, the utilization of liposomes for the delivery of miR-9-5p (miR-9-5p@Lip) presents a promising therapeutic strategy for the treatment of HI injury. Innovation: miR-9-5p is a potential therapeutic agent for ischemic stroke through its modulation of the ZBTB20/Nrf2/Keap1 signaling pathway, influencing mitochondrial function and antioxidant response. Furthermore, the use of liposomal delivery for miR-9-5p offers a promising therapeutic strategy for HI injury. Conclusion: Overexpression of miR-9-5p protects against cerebral HI injury by modulating mitochondrial function through the ZBTB20/Nrf2/Keap1 signaling pathway. Antioxid. Redox Signal. 42, 512-528. [Figure: see text].
Collapse
Affiliation(s)
- Yijing Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Chengcheng Gai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Shuwen Yu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Yan Song
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Bing Gu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Qian Luo
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Xixi Wang
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Quan Hu
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, People's Republic of China
| | - Weiyang Liu
- Jinan Xicheng Experimental High School, Jinan, People's Republic of China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| |
Collapse
|
2
|
Zhang W, Ji X, Zhao Q, Qi J, Guo W, Zhang G, Guan Y, Li S, Mao Y. Nanoodor Particles Deliver Drugs to Central Nervous System via Olfactory Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408908. [PMID: 39998409 PMCID: PMC12005826 DOI: 10.1002/advs.202408908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 02/08/2025] [Indexed: 02/26/2025]
Abstract
Central nervous system (CNS) disorders confront significant challenges in drug delivery due to the blood-brain barrier (BBB). Inspired by the rapid and precise binding of odor molecules to olfactory receptors (ORs), this research uses thiolated HPMA to construct odor nanoparticles (nanoodors) capable of delivering drugs to the CNS via the olfacto-cerebral pathway to overcome the delivery obstruction. The nanoodor core is used to encapsulate agomelatine (AGO), a CNS-targeting antidepressant, and the encapsulation efficiency exceeded 80%. A series of thiol-presenting nanoscale structures with different surface densities of thiol groups are constructed, and the effectiveness positively correlated with the density of thiol groups on their surface. Notably, the nanoodors enable precise brain-targeted delivery, outperforming commercially available oral formulations in terms of drug accumulation in the brain and antidepressant effects. The study of the nanoodor transport and action mechanisms revealed that after binding to ORs, the nanoodors are rapidly delivered to the brain via the olfactory pathway. Nanoodors, the first design to deliver CNS drugs via the olfactory pathway by mimicking natural smells for the treatment of CNS disorders, are expected to achieve clinical transformation, benefiting human health.
Collapse
Affiliation(s)
- Wei Zhang
- Department of PharmaceuticsSchool of PharmacyShenyang Pharmaceutical University103 Wenhua RoadShenyangLiaoning110016P. R. China
| | - Xingwang Ji
- Department of PharmaceuticsSchool of PharmacyShenyang Pharmaceutical University103 Wenhua RoadShenyangLiaoning110016P. R. China
| | - Qianyanqiu Zhao
- Department of PharmaceuticsSchool of PharmacyShenyang Pharmaceutical University103 Wenhua RoadShenyangLiaoning110016P. R. China
| | - Jinyao Qi
- Department of PharmaceuticsSchool of PharmacyShenyang Pharmaceutical University103 Wenhua RoadShenyangLiaoning110016P. R. China
| | - Wen Guo
- Department of PharmaceuticsSchool of PharmacyShenyang Pharmaceutical University103 Wenhua RoadShenyangLiaoning110016P. R. China
| | - Gaoshuo Zhang
- Department of PharmaceuticsSchool of PharmacyShenyang Pharmaceutical University103 Wenhua RoadShenyangLiaoning110016P. R. China
| | - Yujing Guan
- Institute of Cancer MedicineFaculty of MedicineDalian University of TechnologyNo.2 Linggong Road, Ganjingzi DistrictDalianLiaoning116024China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor SurgeryCancer Hospital of Dalian University of TechnologyCancer Hospital of China Medical UniversityLiaoning Cancer Hospital & InstituteShenyangLiaoning110042China
- The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with EngineeringShenyangLiaoning110042China
- Institute of Cancer MedicineFaculty of MedicineDalian University of TechnologyNo.2 Linggong Road, Ganjingzi DistrictDalianLiaoning116024China
| | - Yuling Mao
- Department of PharmaceuticsSchool of PharmacyShenyang Pharmaceutical University103 Wenhua RoadShenyangLiaoning110016P. R. China
| |
Collapse
|
3
|
Aguiar B, Alfenim AR, Machado CS, Moreira J, Pinto M, Otero-Espinar FJ, Borges F, Fernandes C. Exploring Nano-Delivery Systems to Enhance the Edaravone Performance in Amyotrophic Lateral Sclerosis Treatment. Int J Mol Sci 2025; 26:2146. [PMID: 40076771 PMCID: PMC11900301 DOI: 10.3390/ijms26052146] [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: 01/23/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Edaravone is one of the treatment options for Amyotrophic Lateral Sclerosis, but its therapeutic efficacy is limited due to the incapacity to cross the blood-brain barrier, as well as its short life span and poor stability, which is ultimately caused by its tautomerism in physiological condions. This work presents an overview about the use of several nanoformulations based on polymeric, protein, lipidic, or hybrid structure as suitable and stable drug delivery systems for encapsulating edaravone. We also evaluated the functionalization of nanoparticles with pegylated chains using the polyethylene glycol or tocopherol polyethylene glycol succinate and the possibility of preparing polymeric nanoparticles at different pH (7.4, 9, and 11). Edaravone was sucessfully encapsulated in polymeric, lipid-polymer hybrid, and lipidic nanoparticles. The use of higher pH values in the synthesis of polymeric nanoparticles has led to a decrease in nanoparticle size and an increase in the percentage of encapsulation efficiency. However, the resulting nanoformulations are not stable. Only polymeric and hybrid nanoparticles showed good stability over 80 days of storage, mainly at 4 °C. Overall, the nanoformulations tested did not show cytotoxicity in the SH-SY5Y cell line except the nanostructured lipid carrier formulations that showed some cytotoxicity possibly due to lipidic peroxidation. In conclusion, this work shows that edaravone can be encapsulated in different nanocarriers that could act as an interesting alternative for the treatment of Amyotrophic Lateral Sclerosis.
Collapse
Affiliation(s)
- Brandon Aguiar
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, 4169-007 Porto, Portugal; (B.A.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Ana Rita Alfenim
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, 4169-007 Porto, Portugal; (B.A.)
| | - Cláudia Sofia Machado
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, 4169-007 Porto, Portugal; (B.A.)
| | - Joana Moreira
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, 4169-007 Porto, Portugal; (B.A.)
| | - Miguel Pinto
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, 4169-007 Porto, Portugal; (B.A.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Francisco J. Otero-Espinar
- Pharmacology, Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy, Insitute of Materials (iMATUS), University of Santiago de Compostela (USC), 15782 Santiago de Compostela, Spain
- Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain
| | - Fernanda Borges
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, 4169-007 Porto, Portugal; (B.A.)
| | - Carlos Fernandes
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, 4169-007 Porto, Portugal; (B.A.)
| |
Collapse
|
4
|
Baker RS, Wang JTW, Rouatbi N, Lu Y, Al-Adhami T, Asker D, Rahman KM, Al-Chalabi A, Forbes B, Bansal S, Al-Jamal KT. Brain distribution study of [ 14C]-Riluzole following intranasal administration in mice. Int J Pharm 2025; 670:125195. [PMID: 39793633 DOI: 10.1016/j.ijpharm.2025.125195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/14/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) presents a substantial challenge due to its complex nature, limited effective treatment options, and modest benefits from current therapies in slowing disease progression. This study explores the potential of intranasal (IN) delivery to enhance the CNS delivery of riluzole (RLZ), a standard ALS treatment which is subject to blood-brain barrier efflux mechanisms. Additionally, the impact of elacridar (ELC), an efflux pump inhibitor, on IN RLZ CNS bioavailability was examined. To quantify RLZ in vivo in mice, [14C]-RLZ was synthesised using an optimised one-pot method. [14C]-RLZ yield was 21.3 ± 3.4 %, measured by High Performance Liquid Chromatography (HPLC), with a specific activity of 40.4 ± 3.9 µCi/mg measured by HPLC and liquid scintillation counting. RLZ synthesis was verified using proton nuclear magnetic resonance (1H NMR), and liquid chromatography-mass spectrometry. IN RLZ (5 mg/kg) produced double the maximum brain levels (1.11 ± 0.34 % Injected Dose (ID)/brain) at 30 min as oral RLZ (5 mg/kg). The uptake of RLZ in the liver was reduced by half for intranasal administration compared to oral administration. Intravenous ELC (5 mg/kg) substantially increased brain levels of IN RLZ to 3.52 ± 0.62 % ID/g brain at 60 min post-administration, compared to 1.87 ± 0.33 % ID/g brain in the absence of the efflux pump inhibitor. However, increased concentrations were also observed in the liver and blood. These results indicate that intranasal delivery of RLZ enhances brain targeting and reduces liver accumulation compared to the oral route. Brain uptake of IN RLZ was enhanced further by ELC, although not selectively as accumulation in the liver or blood was also observed. Further metabolic research using Chromatography-Mass spectrometry (LC-MS) or NMR along with excretion studies are warranted for a more comprehensive understanding of the pharmacokinetics of IN RLZ and IN RLZ/ELC. Additionally, employing suitable ALS animal models is crucial for understanding RLZ's effects on disease progression, mechanism of action, efficacy, and potential side effects to aid further development.
Collapse
Affiliation(s)
- Rafal S Baker
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Julie Tzu-Wen Wang
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Nadia Rouatbi
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Yuan Lu
- Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang 550004, China
| | - Taha Al-Adhami
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Daniel Asker
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Khondaker Miraz Rahman
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Ben Forbes
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Sukhi Bansal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom; Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China.
| |
Collapse
|
5
|
Yuan M, Han Z, Somayaji Y, Nguyen N, Hu H, Madhu LN, Attaluri S, Kodali M, Yang Y, Hsu YC, Ahuja A, Srinivasan R, Pellois JP, Zhou HC, Shetty AK, Wang Y. Intranasal delivery of metformin using metal-organic framework (MOF)-74-Mg nanocarriers. ADVANCED COMPOSITES AND HYBRID MATERIALS 2025; 8:131. [PMID: 39834534 PMCID: PMC11742004 DOI: 10.1007/s42114-025-01227-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/10/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Dosage tolerance is one of the translational challenges of using metformin (Met) in brain therapeutics. This paper presents metal-organic framework (MOF)-74-Mg nanocarriers (NCs) for intranasal (IN) delivery of brain-specific agents with a prolonged release time. We confirmed their excellent biocompatibility (5 mg/mL) and intrinsic fluorescence properties (370/500 nm excitation/emission peak) in Neuro-2A cells. This NC exhibited a high Met loading rate (10% wt/wt) and a sustained and prolonged release pattern of Met (90% release in 16 h) in Dulbecco's Modified Eagle Medium. We observed an optimal brain accumulation of Met-MOF (9% of the injected dosage) 8 h after IN injection. This percentage is at least 82 times higher than oral administration. Confocal imaging demonstrated significantly higher uptake of Met-MOF, 45 min after IN injection, by 79-85% neurons and 93-97% microglia than astrocytes and oligodendrocytes across 5xFAD mouse brain regions, including hippocampus and striatum. These results suggest MOF-74-Mg is a potential NC for high brain Met accumulation, real-time imaging, and prolonged and sustained release of Met and other neurotherapeutic agents that are challenging to deliver using traditional carriers and administration routes.
Collapse
Affiliation(s)
- Muzhaozi Yuan
- J. Mike Walker ’66 Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843 USA
| | - Zongsu Han
- Department of Chemistry, Texas A&M University, College Station, TX 77843 USA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, College Station, TX 77843 USA
| | - Nguyen Nguyen
- J. Mike Walker ’66 Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843 USA
| | - Hanwen Hu
- J. Mike Walker ’66 Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843 USA
| | - Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, College Station, TX 77843 USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, College Station, TX 77843 USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, College Station, TX 77843 USA
| | - Yihao Yang
- Department of Chemistry, Texas A&M University, College Station, TX 77843 USA
| | - Yu-Chuan Hsu
- Department of Chemistry, Texas A&M University, College Station, TX 77843 USA
| | - Avik Ahuja
- J. Mike Walker ’66 Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843 USA
| | - Rahul Srinivasan
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, 8447 Riverside Pkwy, Bryan, TX 77807 USA
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX 77843 USA
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843 USA
| | - Hong-Cai Zhou
- Department of Chemistry, Texas A&M University, College Station, TX 77843 USA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, College Station, TX 77843 USA
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX 77843 USA
| | - Ya Wang
- J. Mike Walker ’66 Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843 USA
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843 USA
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843 USA
| |
Collapse
|
6
|
Jia CJ, Chen MN, Huang DD, Wu SF, Zeng CM, Liu ZQ, Wang MX, Huang YF, Yuan QJ, Zhang X. Edaravone promotes motoneuron survival and functional recovery after brachial plexus root avulsion and reimplantation in rats: Involvement of SIRT1/TFEB pathway. Int Immunopharmacol 2025; 145:113686. [PMID: 39642559 DOI: 10.1016/j.intimp.2024.113686] [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: 06/09/2024] [Revised: 10/31/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Brachial plexu root avulsion (BPRA) commonly causes extensive motoneuron death, motor axon degeneration and denervation of biceps, leading to devastating motor dysfunction in the upper limb. Edaravone (Eda) has been proven to exert anti-oxidative and neuroprotective effects on various neurological disorders. Herein, we aimed to investigate the efficacy profile and potential mechanisms of Eda on BPRA in vitro and in vivo models. METHODS Rats following BPRA and reimplantation surgery were intraperitoneally injected with Eda once daily. The motor function recovery of the affected forelimb was assessed by Terzis grooming test. Histological staining and transmission electron microscopy were performed to evaluate the morphological appearance of the spinal cord, musculocutaneous nerve, and biceps. Further in-depth studies to explore the underlying mechanisms of Eda were conducted using Western blotting, biochemical assays, and immunofluorescence in H2O2-induced NSC-34 cells. RESULTS Eda significantly accelerated motor function recovery, enhanced motoneuron survival, prevented motor axon descent, preserved myelin sheath integrity and attenuated muscle atrophy. Additionally, Eda treatment markedly suppressed oxidative stress-related indicators, downregulated apoptosis-related proteins, mitigated glial reactivity, and activated SIRT1 and TFEB. Notably, the neuroprotective effect of Eda was diminished by the SIRT1 inhibitor EX527 in H2O2-treated NSC-34 cells, suggesting that Eda regulated oxidative stress and apoptosis through SIRT1/TFEB-induced autophagy flux. CONCLUSIONS Eda enhanced motoneuron survival and axonal regeneration that promotes motor functional restoration by inhibiting oxidative stress and apoptosis via the SIRT1/TFEB-autophagy pathway. Thus, it may serve as a promising strategy in reimplantation surgery for the treatment of BPRA.
Collapse
Affiliation(s)
- Cai-Ju Jia
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Man-Ni Chen
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Dou-Dou Huang
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Shao-Feng Wu
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Chun-Ming Zeng
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Zhe-Qi Liu
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Meng-Xia Wang
- ShunDe Polytechnic College, School of Medicine & Health Care, FoShan, Guangdong, 528000, China.
| | - Yan-Feng Huang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| | - Qiu-Ju Yuan
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Shatin, N.T., Hong Kong SAR, China.
| | - Xie Zhang
- Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, 523808, China.
| |
Collapse
|
7
|
Bagheri Y, Dehghan M, Hejazian SM, Ardalan M, Zununi Vahed S, Niknafs B. The protective effect of Edaravone against acute renocardiac syndrome in a kidney ischemia-reperfusion model. J Cardiovasc Thorac Res 2024; 16:243-248. [PMID: 40027361 PMCID: PMC11866771 DOI: 10.34172/jcvtr.33077] [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: 01/09/2024] [Accepted: 10/20/2024] [Indexed: 03/05/2025] Open
Abstract
Introduction Acute kidney injury (AKI) is a common clinical occurrence causing high mortality and morbidity. In acute renocardiac syndrome, AKI leads to acute cardiac injury or/and dysfunction. This study aimed to investigate the antioxidative effects of Edaravone on cardiac tissues following the induction of renal ischemia-reperfusion injury (IRI) in rats. Methods Twenty-four male Wistar rats were randomly divided into four groups: IR+Edaravone, Edaravone, IR, and Sham groups (six rats per group). Non-traumatic clamps were used to stop the artery and vein blood flow of the left kidney in rats of the IR groups for 45 minutes. Thirty minutes before ischemia induction, Edaravone (3 mg/kg) was injected intraperitoneally in the IR+Edaravone group. Cardiac samples were subjected to biochemical analyses. Results The Results showed a significant increase in the enzymatic activity of glutathione peroxidase (P=0.01), catalase (P=0.03), and superoxide dismutase (P=0.02), and the levels of glutathione (P=0.012), and total antioxidant capacity (P<0.001) in the IR+Edaravone group in comparison to the IR group. Moreover, the total antioxidant capacity of the heart was increased in the Edaravone group compared to the control and IR groups (P<0.001), indicating the safety of the drug. Conclusion The results can reveal important insights into the protective effects of Edaravone against acute renocardiac syndrome.
Collapse
Affiliation(s)
- Yasin Bagheri
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahshid Dehghan
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | - Bahram Niknafs
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Luo X, Luo L, Lai R, Li Y, Zhou H, Li X. Synthesis and Antioxidant Effects of Edaravone-Loaded MPEG-2000-DSPE Micelles in Rotenone-Induced PC12 Cell Model of Parkinson's Disease. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1962. [PMID: 39683350 DOI: 10.3390/nano14231962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder globally that lacks any disease-modifying drug for prevention or treatment. Oxidative stress has been identified as one of the key pathogenic drivers of Parkinson's disease (PD). Edaravone, an approved free-radical scavenger, has proven to have potential against PD by targeting multiple key pathologies, including oxidative stress, focal mitochondria, and neuroinflammation. However, its bioavailability is potentially restricted due to its poor solubility and short half-life. This study aims to develop a simple and effective drug delivery system for edaravone to enhance its solubility, stability, and bioavailability to improve its neuroprotective efficacy. An MPEG-2000-DSPE-edaravone (MDE) micelle was prepared via solvent evaporation using MPEG-2000-DSPE as a carrier to encapsulate edaravone. The morphology, particle size, zeta potential, chemical structure, and edaravone loading of MDE were evaluated. We then investigated whether such targeted edaravone delivery could provide enhanced neuroprotection. A cell model of PD was established in PC12 cells through exposure to rotenone. The effects of MDE on PC12 cells treated with or without rotenone were evaluated using a cell counting kit-8, calcein acetoxymethyl ester (AM)-propidine iodide (PI) staining, and flow cytometry. Cell migration was evaluated using a wound healing assay. Additionally, the intracellular antioxidant study was performed using an ROS-level-detecting DCFH-DA probe, and the mitochondrial membrane potentials were evaluated using a JC-1 assay. MDE with a drug-loading content of 17.6% and an encapsulation efficiency of 92.8% was successfully prepared. The resultant MDE had a mean particle size of 112.97 ± 5.54 nm with a zeta potential of -42 mV. Cytotoxicity assays confirmed that the MDE (≤200 ug/mL) exhibited promising cytocompatibility with no significant effect on cell viability, cell cycle regulation, or apoptosis levels. Likewise, compared with the free edaravone, no effect on cell migration was noted for MDE. MDE might be able to target edaravone delivery into PC12 cells, increasing the mitochondrial membrane potential and providing a significant local antioxidant effect. The results demonstrated that MPEG-2000-DSPE could be a promising material for enhancing edaravone's aqueous solubility, stability, and antioxidant effects. MDE could be a potential drug formulation for treating PD and other diseases in which oxidative stress plays a key role in pathogenesis.
Collapse
Affiliation(s)
- Xin Luo
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Linshan Luo
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Rong Lai
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yan Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Hongyan Zhou
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiting Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| |
Collapse
|
9
|
Duan Z, Zhou W, He S, Wang W, Huang H, Yi L, Zhang R, Chen J, Zan X, You C, Gao X. Intranasal Delivery of Curcumin Nanoparticles Improves Neuroinflammation and Neurological Deficits in Mice with Intracerebral Hemorrhage. SMALL METHODS 2024; 8:e2400304. [PMID: 38577823 DOI: 10.1002/smtd.202400304] [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: 03/02/2024] [Revised: 03/24/2024] [Indexed: 04/06/2024]
Abstract
Intracerebral hemorrhage (ICH) represents one of the most severe subtypes of stroke. Due to the complexity of the brain injury mechanisms following ICH, there are currently no effective treatments to significantly improve patient functional outcomes. Curcumin, as a potential therapeutic agent for ICH, is limited by its poor water solubility and oral bioavailability. In this study, mPEG-PCL is used to encapsulate curcumin, forming curcumin nanoparticles, and utilized the intranasal administration route to directly deliver curcumin nanoparticles from the nasal cavity to the brain. By inhibiting pro-inflammatory neuroinflammation of microglia following ICH in mice, reprogramming pro-inflammatory microglia toward an anti-inflammatory function, and consequently reducing neuronal inflammatory death and hematoma volume, this approach improved blood-brain barrier damage in ICH mice and promoted the recovery of neurological function post-stroke. This study offers a promising therapeutic strategy for ICH to mediate neuroinflammatory microenvironments.
Collapse
Affiliation(s)
- Zhongxin Duan
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Wenjie Zhou
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, 610041, China
| | - Shi He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Wanyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Hongyi Huang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Linbin Yi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Rui Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Junli Chen
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xin Zan
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Chao You
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
10
|
Burban A, Sharanek A, Hernandez-Corchado A, Najafabadi HS, Soleimani VD, Jahani-Asl A. Targeting glioblastoma with a brain-penetrant drug that impairs brain tumor stem cells via NLE1-Notch1 complex. Stem Cell Reports 2024; 19:1534-1547. [PMID: 39423824 PMCID: PMC11589194 DOI: 10.1016/j.stemcr.2024.09.007] [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: 03/08/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024] Open
Abstract
Brain tumor stem cells (BTSCs) are a population of self-renewing malignant stem cells that play an important role in glioblastoma tumor hierarchy and contribute to tumor growth, therapeutic resistance, and tumor relapse. Thus, targeting of BTSCs within the bulk of tumors represents a crucial therapeutic strategy. Here, we report that edaravone is a potent drug that impairs BTSCs in glioblastoma. We show that edaravone inhibits the self-renewal and growth of BTSCs harboring a diverse range of oncogenic mutations without affecting non-oncogenic neural stem cells. Global gene expression analysis revealed that edaravone significantly alters BTSC transcriptome and attenuates the expression of a large panel of genes involved in cell cycle progression, stemness, and DNA repair mechanisms. Mechanistically, we discovered that edaravone directly targets Notchless homolog 1 (NLE1) and impairs Notch signaling pathway, alters the expression of stem cell markers, and sensitizes BTSC response to ionizing radiation (IR)-induced cell death. Importantly, we show that edaravone treatment in preclinical models delays glioblastoma tumorigenesis, sensitizes their response to IR, and prolongs the lifespan of animals. Our data suggest that repurposing of edaravone is a promising therapeutic strategy for patients with glioblastoma.
Collapse
Affiliation(s)
- Audrey Burban
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1E2, Canada; Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Boulevard West, Montréal, QC H4A 3T2, Canada
| | - Ahmad Sharanek
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1E2, Canada; Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Boulevard West, Montréal, QC H4A 3T2, Canada
| | - Aldo Hernandez-Corchado
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montréal, QC H4A 3J1, Canada; Department of Human Genetics, McGill University, 3640 Rue University, Montréal, QC H3A OC7, Canada
| | - Hamed S Najafabadi
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montréal, QC H4A 3J1, Canada; Department of Human Genetics, McGill University, 3640 Rue University, Montréal, QC H3A OC7, Canada
| | - Vahab D Soleimani
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1E2, Canada; Department of Human Genetics, McGill University, 3640 Rue University, Montréal, QC H3A OC7, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1E2, Canada; Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Boulevard West, Montréal, QC H4A 3T2, Canada; Cancer Therapeutic and Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
11
|
Toader C, Radoi MP, Covlea CA, Covache-Busuioc RA, Ilie MM, Glavan LA, Corlatescu AD, Costin HP, Gica MD, Dobrin N. Cerebral Aneurysm: Filling the Gap Between Pathophysiology and Nanocarriers. Int J Mol Sci 2024; 25:11874. [PMID: 39595942 PMCID: PMC11593836 DOI: 10.3390/ijms252211874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Intracranial aneurysms, characterized by abnormal dilations of cerebral arteries, pose significant health risks due to their potential to rupture, leading to subarachnoid hemorrhage with high mortality and morbidity rates. This paper aim is to explore the innovative application of nanoparticles in treating intracranial aneurysms, offering a promising avenue for enhancing current therapeutic strategies. We took into consideration the pathophysiology of cerebral aneurysms, focusing on the role of hemodynamic stress, endothelial dysfunction, and inflammation in their development and progression. By comparing cerebral aneurysms with other types, such as aortic aneurysms, we identify pathophysiological similarities and differences that could guide the adaptation of treatment approaches. The review highlights the potential of nanoparticles to improve drug delivery, targeting, and efficacy while minimizing side effects. We discuss various nanocarriers, including liposomes and polymeric nanoparticles, and their roles in overcoming biological barriers and enhancing therapeutic outcomes. Additionally, we discuss the potential of specific compounds, such as Edaravone and Tanshinone IIA, when used in conjunction with nanocarriers, to provide neuroprotective and anti-inflammatory benefits. By extrapolating insights from studies on aortic aneurysms, new research directions and therapeutic strategies for cerebral aneurysms are proposed. This interdisciplinary approach underscores the potential of nanoparticles to positively influence the management of intracranial aneurysms, paving the way for personalized treatment options that could significantly improve patient outcomes.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Mugurel Petrinel Radoi
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Christian-Adelin Covlea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Milena Monica Ilie
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Luca-Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Antonio-Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Horia-Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Maria-Daria Gica
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | | |
Collapse
|
12
|
Lescouzères L, Patten SA. Promising animal models for amyotrophic lateral sclerosis drug discovery: a comprehensive update. Expert Opin Drug Discov 2024; 19:1213-1233. [PMID: 39115327 DOI: 10.1080/17460441.2024.2387791] [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: 03/08/2024] [Accepted: 07/30/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons. Several animal models have been generated to understand ALS pathogenesis. They have provided valuable insight into disease mechanisms and the development of therapeutic strategies. AREAS COVERED In this review, the authors provide a concise overview of simple genetic model organisms, including C. elegans, Drosophila, zebrafish, and mouse genetic models that have been generated to study ALS. They emphasize the benefits of each model and their application in translational research for discovering new chemicals, gene therapy approaches, and antibody-based strategies for treating ALS. EXPERT OPINION Significant progress is being made in identifying new therapeutic targets for ALS. This progress is being enabled by promising animal models of the disease using increasingly effective genetic and pharmacological strategies. There are still challenges to be overcome in order to achieve improved success rates for translating drugs from animal models to clinics for treating ALS. Several promising future directions include the establishment of novel preclinical protocol standards, as well as the combination of animal models with human induced pluripotent stem cells (iPSCs).
Collapse
Affiliation(s)
- Léa Lescouzères
- INRS - Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Shunmoogum A Patten
- INRS - Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
- Departement de Neurosciences, Université de Montréal, Montreal, Canada
| |
Collapse
|
13
|
Wang G, Zhai Z, Wang W, Xia X, Guo H, Yue X, Wang X, Zhu B, Huang Z, Pan X, Huang Y, Wu C, Zhang X. Tailored Borneol-Modified Lipid Nanoparticles Nasal Spray for Enhanced Nose-to-Brain Delivery to Central Nervous System Diseases. ACS NANO 2024; 18:23684-23701. [PMID: 39158142 DOI: 10.1021/acsnano.4c08279] [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: 08/20/2024]
Abstract
The nanodrug delivery system-based nasal spray (NDDS-NS) can bypass the blood-brain barrier and deliver drugs directly to the brain, offering unparalleled advantages in the treatment of central nervous system (CNS) diseases. However, the current design of NNDS-NS is excessively focused on mucosal absorption while neglecting the impact of nasal deposition on nose-to-brain drug delivery, resulting in an unsatisfactory nose-to-brain delivery efficiency. In this study, the effect of the dispersion medium viscosity on nasal drug deposition and nose-to-brain delivery in NDDS-NS was elucidated. The optimized formulation F5 (39.36 mPa·s) demonstrated significantly higher olfactory deposition fraction (ODF) of 23.58%, and a strong correlation between ODF and intracerebral drug delivery (R2 = 0.7755) was observed. Building upon this understanding, a borneol-modified lipid nanoparticle nasal spray (BLNP-NS) that combined both nasal deposition and mucosal absorption was designed for efficient nose-to-brain delivery. BLNP-NS exhibited an accelerated onset of action and enhanced brain targeting efficiency, which could be attributed to borneol modification facilitating the opening of tight junction channels. Furthermore, BLNP-NS showed superiority in a chronic migraine rat model. It not only provided rapid relief of migraine symptoms but also reversed neuroinflammation-induced hyperalgesia. The results revealed that borneol modification could induce the polarization of microglia, regulate the neuroinflammatory microenvironment, and repair the neuronal damage caused by neuroinflammation. This study highlights the impact of dispersion medium viscosity on the nose-to-brain delivery process of NDDS-NS and serves as a bridge between the formulation development and clinical transformation of NDDS-NS for the treatment of CNS diseases.
Collapse
Affiliation(s)
- Guanlin Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, P. R. China
| | - Zizhao Zhai
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, P. R. China
| | - Wenhao Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, P. R. China
| | - Xiao Xia
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, P. R. China
| | - Haihua Guo
- Guangdong Province Key Laboratory of Utilization and Protection for Resource of Food and Medicinal Plant in North Region, Shaoguan University, Shaoguan 512005, Guangdong, P. R. China
| | - Xiao Yue
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, P. R. China
| | - Xiaoyuan Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, P. R. China
| | - Bing Zhu
- Respirent Pharmaceuticals, Co., Ltd., Chongqing 400714, P. R. China
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, P. R. China
- Institute of Advanced Drug Delivery Systems, Jinan University, Guangzhou 510006, Guangdong, P. R. China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, P. R. China
| | - Ying Huang
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, P. R. China
- Institute of Advanced Drug Delivery Systems, Jinan University, Guangzhou 510006, Guangdong, P. R. China
| | - Chuanbin Wu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, P. R. China
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, P. R. China
- Institute of Advanced Drug Delivery Systems, Jinan University, Guangzhou 510006, Guangdong, P. R. China
| | - Xuejuan Zhang
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, P. R. China
- Institute of Advanced Drug Delivery Systems, Jinan University, Guangzhou 510006, Guangdong, P. R. China
| |
Collapse
|
14
|
Arayici PP, Coksu I, Ozbek T, Acar S. Targeted delivery of rifaximin using P6.2-decorated bifunctional PLGA nanoparticles for combating Staphylococcus aureus infections. BIOMATERIALS ADVANCES 2024; 161:213862. [PMID: 38678666 DOI: 10.1016/j.bioadv.2024.213862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/02/2024] [Accepted: 04/13/2024] [Indexed: 05/01/2024]
Abstract
The emergence of antibiotic resistance makes the treatment of bacterial infections difficult and necessitates the development of alternative strategies. Targeted drug delivery systems are attracting great interest in overcoming the limitations of traditional antibiotics. Here, we aimed for targeted delivery of rifaximin (RFX) by decorating RFX-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) with synthetic P6.2 peptide, which was used as a targeting agent for the first time. Our results showed that encapsulation of RFX into NPs increased its antibacterial activity by improving its solubility and providing controlled release, while P6.2 modification allowed targeting of NPs to S. aureus bacterial cells. A promising therapeutic approach for bacterial infections, these P6.2-conjugated RFX-loaded PLGA NPs (TR-NP) demonstrated potent antibacterial activity against both strains of S. aureus. The antibacterial activity of RFX-loaded PLGA NPs (R-NP) showed significant results with an increase of 8 and 16-fold compared to free RFX against S. aureus and MRSA, respectively. Moreover, the activity of targeted nanoparticles was found to be increased 32 or 16-fold with an MBC value of 0.0078 μg/mL. All nanoparticles were found to be biocompatible at doses where they showed antimicrobial activity. Finally, it revealed that P6.2-conjugated targeted nanoparticles extremely accumulated in S. aureus rather than E. coli.
Collapse
Affiliation(s)
- Pelin Pelit Arayici
- Yildiz Technical University, Faculty of Chemical and Metallurgical, Department of Bioengineering, 34210 Esenler, Istanbul, Turkey; Health Biotechnology Joint Research and Application Center of Excellence, 34220 Esenler, Istanbul, Turkey.
| | - Irem Coksu
- Yildiz Technical University, Faculty of Chemical and Metallurgical, Department of Bioengineering, 34210 Esenler, Istanbul, Turkey
| | - Tulin Ozbek
- Yildiz Technical University, Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, 34220 Esenler, Istanbul, Turkey
| | - Serap Acar
- Yildiz Technical University, Faculty of Chemical and Metallurgical, Department of Bioengineering, 34210 Esenler, Istanbul, Turkey
| |
Collapse
|
15
|
Meng YQ, Ren J, Sun JX, Guo FY, Min JZ, Nan JX, Quan JS, Lian LH, Jin CH. Synthesis and anti-liver fibrosis activity of imidazole and thiazole compounds containing amino acids. Eur J Med Chem 2024; 269:116311. [PMID: 38508118 DOI: 10.1016/j.ejmech.2024.116311] [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: 10/14/2023] [Revised: 01/27/2024] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
Four series of imidazoles (15a-g, 20c, and 20d) and thiazoles (18a-g, 22a, and 22b) possessing various amino acids were synthesized and evaluated for activin receptor-like kinase 5 (ALK5) inhibitory activities in an enzymatic assay. Among them, compounds 15g and 18c showed the highest inhibitory activity against ALK5, with IC50 values of 0.017 and 0.025 μM, respectively. Compounds 15g and 18c efficiently inhibited extracellular matrix (ECM) deposition in TGF-β-induced hepatic stellate cells (HSCs), and eventually suppressed HSC activation. Moreover, compound 15g showed a good pharmacokinetic (PK) profile with a favorable half-life (t1/2 = 9.14 h). The results indicated that these compounds exhibited activity targeting ALK5 and may have potential in the treatment of liver fibrosis; thus they are worthy of further study.
Collapse
Affiliation(s)
- Yu-Qing Meng
- Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China
| | - Jie Ren
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Jing-Xin Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Fang-Yan Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Jun-Zhe Min
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Ji-Xing Nan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China; Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China
| | - Ji-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China; Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China.
| | - Li-Hua Lian
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China; Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China.
| | - Cheng-Hua Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, China; Interdisciplinary Program of Biological Function Molecules, College of Integration Science, Yanbian University, Yanji, 133002, China.
| |
Collapse
|
16
|
Zhou Y, Guo L, Dai G, Li B, Bai Y, Wang W, Chen S, Zhang J. An Overview of Polymeric Nanoplatforms to Deliver Veterinary Antimicrobials. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:341. [PMID: 38392714 PMCID: PMC10893358 DOI: 10.3390/nano14040341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 02/24/2024]
Abstract
There is an urgent need to find new solutions for the global dilemma of increasing antibiotic resistance in humans and animals. Modifying the performance of existing antibiotics using the nanocarrier drug delivery system (DDS) is a good option considering economic costs, labor costs, and time investment compared to the development of new antibiotics. Numerous studies on nanomedicine carriers that can be used for humans are available in the literature, but relatively few studies have been reported specifically for veterinary pharmaceutical products. Polymer-based nano-DDS are becoming a research hotspot in the pharmaceutical industry owing to their advantages, such as stability and modifiability. This review presents current research progress on polymer-based nanodelivery systems for veterinary antimicrobial drugs, focusing on the role of polymeric materials in enhancing drug performance. The use of polymer-based nanoformulations improves treatment compliance in livestock and companion animals, thereby reducing the workload of managers. Although promising advances have been made, many obstacles remain to be addressed before nanoformulations can be used in a clinical setting. Some crucial issues currently facing this field, including toxicity, quality control, and mass production, are discussed in this review. With the continuous optimization of nanotechnology, polymer-based DDS has shown its potential in reducing antibiotic resistance to veterinary medicines.
Collapse
Affiliation(s)
- Yaxin Zhou
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou 730050, China; (Y.Z.); (G.D.); (B.L.); (Y.B.); (W.W.)
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou 730050, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Lihua Guo
- Shenniu Pharmaceutical Co., Ltd., Dezhou 253034, China;
| | - Guonian Dai
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou 730050, China; (Y.Z.); (G.D.); (B.L.); (Y.B.); (W.W.)
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou 730050, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Bing Li
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou 730050, China; (Y.Z.); (G.D.); (B.L.); (Y.B.); (W.W.)
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou 730050, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Yubin Bai
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou 730050, China; (Y.Z.); (G.D.); (B.L.); (Y.B.); (W.W.)
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou 730050, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Weiwei Wang
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou 730050, China; (Y.Z.); (G.D.); (B.L.); (Y.B.); (W.W.)
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou 730050, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Shulin Chen
- College of Veterinary Medicine, Northwest A & F University, Yangling 712100, China
| | - Jiyu Zhang
- Key Laboratory of New Animal Drug Project of Gansu Province, Lanzhou 730050, China; (Y.Z.); (G.D.); (B.L.); (Y.B.); (W.W.)
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou 730050, China
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| |
Collapse
|
17
|
Huang Q, Chen Y, Zhang W, Xia X, Li H, Qin M, Gao H. Nanotechnology for enhanced nose-to-brain drug delivery in treating neurological diseases. J Control Release 2024; 366:519-534. [PMID: 38182059 DOI: 10.1016/j.jconrel.2023.12.054] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/07/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Despite the increasing global incidence of brain disorders, achieving sufficient delivery towards the central nervous system (CNS) remains a formidable challenge in terms of translating into improved clinical outcomes. The brain is highly safeguarded by physiological barriers, primarily the blood-brain barrier (BBB), which routinely excludes most therapeutics from entering the brain following systemic administration. Among various strategies investigated to circumvent this challenge, intranasal administration, a noninvasive method that bypasses the BBB to allow direct access of drugs to the CNS, has been showing promising results. Nanotechnology-based drug delivery systems, in particular, have demonstrated remarkable capacities in overcoming the challenges posed by nose-to-brain drug delivery and facilitating targeted drug accumulation within the brain while minimizing side effects of systemic distribution. This review comprehensively summarizes the barriers of nose-to-brain drug delivery, aiming to enhance our understanding of potential physiological obstacles and improve the efficacy of nasal delivery in future trials. We then highlight cutting-edge nanotechnology-based studies that enhance nose-to-brain drug delivery in three key aspects, demonstrating substantial potential for improved treatment of brain diseases. Furthermore, the attention towards clinical studies will ease the regulatory approval process for nasal administration of nanomedicines targeting brain disease.
Collapse
Affiliation(s)
- Qianqian Huang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yongke Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Weiwei Zhang
- Department of Public Health, Chengdu Medical College, 783 Xindu Avenue, Xindu, Chengdu, Sichuan 610500, China
| | - Xue Xia
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Hanmei Li
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Meng Qin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610064, China.
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610064, China.
| |
Collapse
|