1
|
Rustage K, Rai N, Sinha SK, Goyal J, Chouhan P, Baniya B, Dubey D, Singhal R, Malani P, Pareek A, Pant M, Jain S, Bisht A, Pareek A, Ratan Y, Ashraf GM, Jain V. Evaluation of the Sporadic Anti-Alzheimer's Activity of Purpurin Using In Silico, In Vitro, and In Vivo Approaches. Mol Neurobiol 2025:10.1007/s12035-025-04910-9. [PMID: 40210836 DOI: 10.1007/s12035-025-04910-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
Purpurin, a naturally occurring compound found in certain plants, has demonstrated promising neuroprotective effects in the context of Alzheimer's disease (AD). This study investigated the efficacy of purpurin in mitigating neurodegenerative changes induced by streptozotocin (3 mg/kg ICV) and amyloid beta (20 μM) in murine models. Neuroprotective effects were assessed through in vitro and in vivo experiments complemented by in silico simulation studies. SH-SY5Y cell viability, behavioral, biochemical, and histopathological studies were also conducted. The results revealed that purpurin interacts with acetylcholinesterase (AChE) and amyloid-beta (Aβ), exhibiting glide scores of - 10.72 and - 3.05 kcal/mol, respectively. Purpurin (8 μM) significantly alleviated Aβ-induced cellular damage by decreasing malondialdehyde production and enhancing superoxide dismutase and Thio barbituric acid reactive substances levels in a dose-dependent manner. Intraperitoneal administration of purpurin at 50 mg/kg significantly improved both long-term and short-term memory and enhanced social interactions. These benefits were linked to the reductions in AChE activity and oxidative and inflammatory marker levels triggered by streptozotocin. Neuroprotective effects were also supported by restoring neuronal DNA content in the hippocampus, cerebellum and prefrontal cortex. Histological findings further corroborated the reduction in neurodegenerative marker levels. In silico simulations supported these findings by indicating that purpurin primarily binds to the Trp 286 and Tyr 341 residues of AChE, inhibiting its catalytic activity at the peripheral anionic site. In conclusion, the neuroprotective activity of purpurin in AD models is attributed to its inhibitory effects on AChE, coupled with reductions in inflammation and oxidative stress and the restoration of neuronal DNA integrity in critical brain regions.
Collapse
Affiliation(s)
- Kajol Rustage
- Department of Pharmaceutical Sciences, Mohan Lal Sukhadia University, Udaipur, 313001, India
| | - Nitish Rai
- Department of Biotechnology, Mohan Lal Sukhadia University, Udaipur, 313001, India
| | - Saurabh Kumar Sinha
- Department of Pharmaceutical Sciences, Mohan Lal Sukhadia University, Udaipur, 313001, India
| | - Juhi Goyal
- Department of Biotechnology, Mohan Lal Sukhadia University, Udaipur, 313001, India
| | - Pragati Chouhan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India
| | - Bhuvanesh Baniya
- Department of Pharmaceutical Sciences, Mohan Lal Sukhadia University, Udaipur, 313001, India
| | - Deepti Dubey
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India
| | - Runjhun Singhal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India
| | - Pooja Malani
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India
| | - Malvika Pant
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India
| | - Smita Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India
| | - Akansha Bisht
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India
| | - Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India.
| | - Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India
| | - Ghulam Md Ashraf
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, SIP, Suzhou, Jiangsu Province, 215123, People's Republic of China
| | - Vivek Jain
- Department of Pharmaceutical Sciences, Mohan Lal Sukhadia University, Udaipur, 313001, India.
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, India.
| |
Collapse
|
2
|
Zhou LY, Wang XB, Chen XQ, Li R, Yu BB, Pan MX, Fang L, Li J, Cui XJ, Yao M, Lu X. Neuroprotective effect and possible mechanism of edaravone in rat models of spinal cord injury: a systematic review and network meta-analysis. Front Pharmacol 2025; 16:1538879. [PMID: 40260386 PMCID: PMC12009846 DOI: 10.3389/fphar.2025.1538879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/06/2025] [Indexed: 04/23/2025] Open
Abstract
Objective The present review was developed to critically evaluate the neuroprotective effects of edaravone for experimental rat models of spinal cord injury (SCI) and generalize the possible mechanisms. Methods Systematic searches were carried out on databases including PubMed, Embase, Web of Science, Scopus, and Cochrane Library from their inception to March 2024. Controlled studies that assessed the neurological roles of edaravone on rats following SCI were selected. The Basso, Beattie, and Bresnahan (BBB) locomotor rating scale, residual white matter area, and malondialdehyde (MDA) level of the SCI rats were systematically searched by two reviewers. Results Ten eligible publications were included. Meta-analyses showed increased BBB scores in edaravone-treated rats compared with control ones. The effect size gradually increased from day 7 (seven studies, n = 246, weighted mean difference (WMD) = 1.96, 95% confidence interval (CI) = 1.23 to 2.68, P < 0.00001) to day 28 (seven studies, n = 222, WMD = 4.41, 95% CI = 3.19 to 5.63, P < 0.00001) after injury and then maintained stably in the following time. Meanwhile, edaravone treatment was associated with an amendment in the spared area of white matter and a lowering in the MDA expression in the lesion area. The subgroup analyses revealed that rats treated with edaravone exhibited superior locomotor recovery in compression injury models than contusion ones. In network analyses, the surface under the cumulative ranking curve gradually increased up to a dose of 5-6 mg/(kg·d) of edaravone, after which it plateaued. Mechanism analysis suggested edaravone can ameliorate oxidative stress, mitigate neuroinflammation, and counteract neuron apoptosis and ferroptosis via multiple signaling pathways to exert its neuroprotective effects. Conclusion Collectively, with a protective effect and a systematic action mechanism, edaravone warrants further investigation in SCI research and treatment. Nonetheless, in light of the limitations in the included studies, the findings in this review should be interpreted with caution. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/view/CRD42022374914.
Collapse
Affiliation(s)
- Long-yun Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xiao-bo Wang
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu-qing Chen
- Department of Otolaryngology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ran Li
- Department of Rehabilitation Medicine, Traditional Chinese Medicine Hospital of LuAn, Luan, China
| | - Bin-bin Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Meng-xiao Pan
- Department of Rehabilitation Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Lu Fang
- Department of Rehabilitation Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jian Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xue-jun Cui
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Yao
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Lu
- Department of Rehabilitation Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Yang L, Zhu L, Lin B, Shi Y, Lai W, Li K, Tian L, Xi Z, Liu H. CuO-NPs Induce Apoptosis and Functional Impairment in BV2 Cells Through the CSF-1R/PLCγ2/ERK/Nrf2 Pathway. TOXICS 2025; 13:231. [PMID: 40278547 PMCID: PMC12031120 DOI: 10.3390/toxics13040231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 04/26/2025]
Abstract
Copper oxide nanoparticles (CuO-NPs) induce neurological diseases, including neurobehavioral defects and neurodegenerative diseases. Direct evidence indicates that CuO-NPs induce inflammation in the central nervous system and cause severe neurotoxicity. However, the mechanism of CuO-NP-induced damage to the nervous system has rarely been studied, and the toxicity of different CuO-NP particle sizes and their copper ion (Cu2+) precipitation in microglia (BV2 cells) is worth exploring. Therefore, this study investigated CuO-NPs with different particle sizes (small particle size: S-CuO-NPs; large particle size: L-CuO-NPs), Cu2+ with equal molar mass (replaced by CuCl2 [Equ group]), and Cu2+ precipitated in a cell culture solution with CuO-NPs (replaced by CuCl2 [Pre group]), and examined the mechanism of action of each on BV2 microglia after co-culture for 12 h and 24 h. The activity of BV2 cells decreased, the morphology was damaged, and the apoptosis rate increased in all the exposed groups. Toxicity increased time- and dose-dependently, and was highest in the Equ group, followed by the S-CuO-NPs, L-CuO-NPs, and Pre groups, respectively. Subsequently, we investigated the mechanism of S-CuO-NP-induced cell injury, and revealed that S-CuO-NPs induced oxidative stress and inflammatory response and increased the membrane permeability of BV2 cells. Moreover, S-CuO-NPs reduced the ratio of p-CSF-1R/CSF-1R, p-PLCγ2/PLCγ2, p-extracellular signal-regulated kinase (ERK)/ERK, p-Nrf2/Nrf2, and Bcl-2/Bax protein expression in microglia, and elevated cleaved caspase-3 expression. The CSF-1R/PLCγ2/ERK/Nrf2 apoptotic pathway was activated. The downregulation of CX3CR1, CSF-1R, brain-derived neurotrophic factor (BDNF), and IGF-1 protein expression indicates impairment of the repair and protection functions of microglia in the nervous system. In summary, our results reveal that CuO-NPs promote an increase in inflammatory molecules in BV2 microglia through oxidative stress, activate the CSF-1R/PLCγ2/ERK/Nrf2 pathway, cause apoptosis, and ultimately result in neurofunctional damage to microglia.
Collapse
Affiliation(s)
- Linhui Yang
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai 201306, China; (L.Y.); (L.Z.)
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin 300050, China; (B.L.); (Y.S.); (W.L.); (K.L.); (L.T.)
| | - Lina Zhu
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai 201306, China; (L.Y.); (L.Z.)
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin 300050, China; (B.L.); (Y.S.); (W.L.); (K.L.); (L.T.)
| | - Bencheng Lin
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin 300050, China; (B.L.); (Y.S.); (W.L.); (K.L.); (L.T.)
| | - Yue Shi
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin 300050, China; (B.L.); (Y.S.); (W.L.); (K.L.); (L.T.)
| | - Wenqing Lai
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin 300050, China; (B.L.); (Y.S.); (W.L.); (K.L.); (L.T.)
| | - Kang Li
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin 300050, China; (B.L.); (Y.S.); (W.L.); (K.L.); (L.T.)
| | - Lei Tian
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin 300050, China; (B.L.); (Y.S.); (W.L.); (K.L.); (L.T.)
| | - Zhuge Xi
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin 300050, China; (B.L.); (Y.S.); (W.L.); (K.L.); (L.T.)
| | - Huanliang Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin 300050, China; (B.L.); (Y.S.); (W.L.); (K.L.); (L.T.)
| |
Collapse
|
4
|
Pal C. Mitochondria-targeting by small molecules against Alzheimer's disease: A mechanistic perspective. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167617. [PMID: 39647244 DOI: 10.1016/j.bbadis.2024.167617] [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: 08/06/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
Alzheimer's disease (AD) poses a considerable worldwide health obstacle, marked by gradual cognitive deterioration and neuronal loss. While the molecular mechanisms underlying AD pathology have been elucidated to some extent, therapeutic options remain limited. Mitochondrial dysfunction has become recognized as a significant factor in the development of AD, with oxidative stress and disrupted energy metabolism being critical elements. This review explores the mechanistic aspects of small molecule targeting of mitochondria as a potential therapeutic approach for AD. The review explores the role of mitochondrial dysfunction in AD, including its involvement in the accumulation of β-amyloid plaques and neurofibrillary tangles, synaptic dysfunction, and neuronal death. Furthermore, the effects of oxidative stress on mitochondrial function were investigated, including the resulting damage to mitochondrial components. Mitochondrial-targeted therapies have attracted attention for their potential to restore mitochondrial function and reduce AD pathology. The review outlines the latest preclinical and clinical evidence supporting the effectiveness of small molecules in targeting mitochondrial dysfunction in AD. Additionally, it discusses the molecular pathways involved in mitochondrial dysfunction and examines how small molecules can intervene to address these abnormalities. By providing a comprehensive overview of the latest research in this field, this review aims to shed light on the therapeutic potential of small molecule targeting of mitochondria in AD and stimulate further research in this promising area of drug development.
Collapse
Affiliation(s)
- Chinmay Pal
- Department of Chemistry, Gobardanga Hindu College, North 24 Parganas, West Bengal 743273, India.
| |
Collapse
|
5
|
Ju JJ, Hang LH. Neuroinflammation and iron metabolism after intracerebral hemorrhage: a glial cell perspective. Front Neurol 2025; 15:1510039. [PMID: 39882361 PMCID: PMC11774705 DOI: 10.3389/fneur.2024.1510039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is the most common subtype of hemorrhagic stroke causing significant morbidity and mortality. Previously clinical treatments for ICH have largely been based on a single pathophysiological perspective, and there remains a lack of curative interventions. Following the rupture of cerebral blood vessels, blood metabolites activate resident immune cells such as microglia and astrocytes, and infiltrate peripheral immune cells, leading to the release of a series of inflammatory mediators. Degradation of hemoglobin produces large amounts of iron ions, leading to an imbalance of iron homeostasis and the production of large quantities of harmful hydroxyl radicals. Neuroinflammation and dysregulation of brain iron metabolism are both important pathophysiological changes in ICH, and both can exacerbate secondary brain injury. There is an inseparable relationship between brain iron metabolism disorder and activated glial cells after ICH. Glial cells participate in brain iron metabolism through various mechanisms; meanwhile, iron accumulation exacerbates neuroinflammation by activating inflammatory signaling pathways modulating the functions of inflammatory cells, and so on. This review aims to explore neuroinflammation from the perspective of iron metabolism, linking the complex pathophysiological changes, delving into the exploration of treatment approaches for ICH, and offering insights that could enhance clinical management strategies.
Collapse
Affiliation(s)
- Jia-Jun Ju
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, China
| | - Li-Hua Hang
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, China
- Kunshan Cancer Pain Prevention and Treatment Key Laboratory, Kunshan, China
| |
Collapse
|
6
|
Rahmati-Dehkordi F, Khanifar H, Zare-Hoseinabadi A, Dadgostar E, Jafarpour H, Aschner M, Mirzaei H, Tamtaji OR, Nabavizadeh F. Potential of Edaravone Dexborneol in the treatment of cerebral ischemia: focus on cell death-related signaling pathways. Mol Biol Rep 2024; 51:1007. [PMID: 39312062 DOI: 10.1007/s11033-024-09952-1] [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: 06/13/2024] [Accepted: 09/16/2024] [Indexed: 02/06/2025]
Abstract
Cerebral ischemia has the highest global rate of morbidity and mortality. It occurs when a sudden occlusion develops in the arterial system, and consequently some parts of the brain are deprived from glucose and oxygen due to the cessation of blood flow. The ensuing reperfusion of the ischemic area results in a cascade of pathological alternations like neuronal apoptosis by producing excessive reactive oxygen species (ROS), oxidative stress and neuroinflammation. Edaravone Dexborneol is a novel agent, comprised of Edaravone and Dexborneol in a 4:1 ratio. It has documented neuroprotective effects against cerebral ischemia injury. Edaravone Dexborneol improves neurobehavioral and sensorimotor function, cognitive function, brain edema, and blood-brain barrier (BBB) integrity in experimental models. It at dosages ranging between 0.375 and 15 mg/kg (from immediately after ischemia until the 28th post-ischemic days) has shown neuroprotective effects in experimental models of cerebral ischemia by inhibiting cell death-signaling pathways. For example, it inhibits apoptosis by increasing Bcl2, and reducing Bax and caspase-3 expression. Edaravone Dexborneol also inhibits pyroptosis by attenuating NF-κB/NLRP3/GSDMD signaling, as well as ferroptosis by activating the Nrf-2/HO-1/GPX4 signaling pathway. It also inhibits autophagy by targeting PI3K/Akt/mTOR signaling pathway. Here, we provide a review on the impacts of Edaravone Dexborneol on cerebral ischemia.
Collapse
Affiliation(s)
- Fatemeh Rahmati-Dehkordi
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Hadi Khanifar
- Department of Internal Medicine, Shahrekord University of Medical Sciences, Shahrekord, Islamic Republic of Iran
| | - Alireza Zare-Hoseinabadi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran
| | - Ehsan Dadgostar
- Behavioral Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran
| | - Hamed Jafarpour
- Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Islamic Republic of Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Omid Reza Tamtaji
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Fatemeh Nabavizadeh
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
| |
Collapse
|
7
|
Hou Y, Shang Y, Xu F, Li T, Li M, Wei L, Fan S, Hou W, Gou W, Shang H, Li Y. Ionizing radiation induces neurotoxicity in Xenopus laevis embryos through neuroactive ligand-receptor interaction pathway. ENVIRONMENTAL RESEARCH 2024; 256:119237. [PMID: 38810829 DOI: 10.1016/j.envres.2024.119237] [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: 02/01/2024] [Revised: 05/14/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Ionizing radiation (IR) poses a significant threat to both the natural environment and biological health. Exposure to specific doses of ionizing radiation early in an organism's development can lead to developmental toxicity, particularly neurotoxicity. Through experimentation with Xenopus laevis (X. laevis), we examined the effects of radiation on early developmental stage. Our findings revealed that radiation led to developmental abnormalities and mortality in X. laevis embryos in a dose-dependent manner, disrupting redox homeostasis and inducing cell apoptosis. Additionally, radiation caused neurotoxic effects, resulting in abnormal behavior and neuron damage in the embryos. Further investigation into the underlying mechanisms of radiation-induced neurotoxicity indicated the potential involvement of the neuroactive ligand-receptor interaction pathway, which was supported by RNA-Seq analysis. Validation of gene expression associated with this pathway and analysis of neurotransmitter levels confirmed our hypothesis. In addition, we further validated the important role of this signaling pathway in radiation-induced neurotoxicity through edaravone rescue experiments. This research establishes a valuable model for radiation damage studying and provides some insight into radiation-induced neurotoxicity mechanisms.
Collapse
Affiliation(s)
- Yue Hou
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Yue Shang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Feifei Xu
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Tingyang Li
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Min Li
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Ling Wei
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, 510006, Guangzhou, China
| | - Saijun Fan
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Wenbin Hou
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China
| | - Wenfeng Gou
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China.
| | - Haihua Shang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China.
| | - Yiliang Li
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, China.
| |
Collapse
|
8
|
Lu H, Chen M, Zhu C. Intranasal Administration of Apelin-13 Ameliorates Cognitive Deficit in Streptozotocin-Induced Alzheimer's Disease Model via Enhancement of Nrf2-HO1 Pathways. Brain Sci 2024; 14:488. [PMID: 38790466 PMCID: PMC11118954 DOI: 10.3390/brainsci14050488] [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: 04/10/2024] [Revised: 05/01/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND The discovery of novel diagnostic methods and therapies for Alzheimer's disease (AD) faces significant challenges. Previous research has shed light on the neuroprotective properties of Apelin-13 in neurodegenerative disorders. However, elucidating the mechanism underlying its efficacy in combating AD-related nerve injury is imperative. In this study, we aimed to investigate Apelin-13's mechanism of action in an in vivo model of AD induced by streptozocin (STZ). METHODS We utilized an STZ-induced nerve injury model of AD in mice to investigate the effects of Apelin-13 administration. Apelin-13 was administered intranasally, and cognitive impairment was assessed using standardized behavioral tests, primarily, behavioral assessment, histological analysis, and biochemical assays, in order to evaluate synaptic plasticity and oxidative stress signaling pathways. RESULTS Our findings indicate that intranasal administration of Apelin-13 ameliorated cognitive impairment in the STZ-induced AD model. Furthermore, we observed that this effect was potentially mediated by the enhancement of synaptic plasticity and the attenuation of oxidative stress signaling pathways. CONCLUSIONS The results of this study suggest that intranasal administration of Apelin-13 holds promise as a therapeutic strategy for preventing neurodegenerative diseases such as AD. By improving synaptic plasticity and mitigating oxidative stress, Apelin-13 may offer a novel approach to neuroprotection in AD and related conditions.
Collapse
Affiliation(s)
- Hai Lu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Fudan University, Shanghai 200032, China; (H.L.); (M.C.)
- College of Clinical Medicine, Jining Medical University, Jining 272067, China
| | - Ming Chen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Fudan University, Shanghai 200032, China; (H.L.); (M.C.)
| | - Cuiqing Zhu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, Fudan University, Shanghai 200032, China; (H.L.); (M.C.)
| |
Collapse
|
9
|
Veroni C, Olla S, Brignone MS, Siguri C, Formato A, Marra M, Manzoli R, Macario MC, Ambrosini E, Moro E, Agresti C. The Antioxidant Drug Edaravone Binds to the Aryl Hydrocarbon Receptor (AHR) and Promotes the Downstream Signaling Pathway Activation. Biomolecules 2024; 14:443. [PMID: 38672460 PMCID: PMC11047889 DOI: 10.3390/biom14040443] [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: 01/17/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
A considerable effort has been spent in the past decades to develop targeted therapies for the treatment of demyelinating diseases, such as multiple sclerosis (MS). Among drugs with free radical scavenging activity and oligodendrocyte protecting effects, Edaravone (Radicava) has recently received increasing attention because of being able to enhance remyelination in experimental in vitro and in vivo disease models. While its beneficial effects are greatly supported by experimental evidence, there is a current paucity of information regarding its mechanism of action and main molecular targets. By using high-throughput RNA-seq and biochemical experiments in murine oligodendrocyte progenitors and SH-SY5Y neuroblastoma cells combined with molecular docking and molecular dynamics simulation, we here provide evidence that Edaravone triggers the activation of aryl hydrocarbon receptor (AHR) signaling by eliciting AHR nuclear translocation and the transcriptional-mediated induction of key cytoprotective gene expression. We also show that an Edaravone-dependent AHR signaling transduction occurs in the zebrafish experimental model, associated with a downstream upregulation of the NRF2 signaling pathway. We finally demonstrate that its rapid cytoprotective and antioxidant actions boost increased expression of the promyelinating Olig2 protein as well as of an Olig2:GFP transgene in vivo. We therefore shed light on a still undescribed potential mechanism of action for this drug, providing further support to its therapeutic potential in the context of debilitating demyelinating conditions.
Collapse
Affiliation(s)
- Caterina Veroni
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.V.); (M.S.B.); (E.A.)
| | - Stefania Olla
- Institute for Genetic and Biomedical Research (IRGB), The National Research Council (CNR), Monserrato, 09042 Cagliari, Italy; (S.O.); (C.S.)
| | - Maria Stefania Brignone
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.V.); (M.S.B.); (E.A.)
| | - Chiara Siguri
- Institute for Genetic and Biomedical Research (IRGB), The National Research Council (CNR), Monserrato, 09042 Cagliari, Italy; (S.O.); (C.S.)
| | - Alessia Formato
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Campus Adriano Buzzati Traverso, Monterotondo Scalo, 00015 Rome, Italy;
| | - Manuela Marra
- Core Facilities Technical-Scientific Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Rosa Manzoli
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (R.M.); (M.C.M.)
| | - Maria Carla Macario
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (R.M.); (M.C.M.)
- Department of Biology, University of Padova, 35121 Padova, Italy
| | - Elena Ambrosini
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.V.); (M.S.B.); (E.A.)
| | - Enrico Moro
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (R.M.); (M.C.M.)
| | - Cristina Agresti
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.V.); (M.S.B.); (E.A.)
| |
Collapse
|
10
|
Montazeri S, Bijani S, Anoush M, Sharafi A, Kalantari-Hesari A, Hosseini MJ. Edaravone Improved Behavioral Abnormalities, Alleviated Oxidative Stress Inflammation, and Metabolic Homeostasis Pathways in Depression. Depress Anxiety 2023; 2023:6623141. [PMID: 40224598 PMCID: PMC11921851 DOI: 10.1155/2023/6623141] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/17/2023] [Accepted: 06/28/2023] [Indexed: 04/15/2025] Open
Abstract
Depression is one of the main factors affecting our daily performance. Among many putative compounds with effect on behavioral and pathophysiological alterations in depression, edaravone (EDV) demonstrates antioxidant and free radical scavenging properties. To investigate possible antidepressive and anxiolytic-like effects of EDV, Wistar rats were randomly divided into six groups: (1) control; (2) EDV (6 mg/kg); (3) post weaning social isolation (PWSI); (4) PWSI+EDV (1.5 mg/kg); (5) PWSI+EDV (3 mg/kg); and (6) PWSI+EDV (6 mg/kg). After the series of behavioral tests, animals were sacrificed, and their hippocampi were dissected for further biochemical and gene expression assays. Our results showed that treatment with 3 and 6 mg/kg EDV after social isolation would improve anxiety, depressive and anhedonic-like behavior in OFT, EPM, FST, and splash tests. In addition, treatment at the aforementioned doses achieved to recover total cellular antioxidant and GSH level. These effects were accompanied with the suppressive effect of EDV on MDA and PCO levels. EDV treatment also modulated the expression of AMPK, Tlr-4, BDNF, nNOS, and iNOS genes. The treatment with 3 and 6 mg/kg EDV would lead to the recovery of behavioral impairments, cellular free radical surge that could be in correlation with the effect of this substance on immune system response, improved energy production system, and more efficacy in the recovery of neural tissue. In conclusion, EDV ameliorates depressive-like disorder by modulating neuroinflammation, energy production, and neural tissue recovery.
Collapse
Affiliation(s)
- Sina Montazeri
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Soroush Bijani
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahdieh Anoush
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Sharafi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Kalantari-Hesari
- Department of Pathobiology, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
| | - Mir-Jamal Hosseini
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
11
|
Zhu M, Zhang Y, Zhang C, Chen L, Kuang Y. Rutin modified selenium nanoparticles reduces cell oxidative damage induced by H 2O 2 by activating Nrf2/HO-1 signaling pathway. J Biomater Appl 2023:8853282231182765. [PMID: 37285508 DOI: 10.1177/08853282231182765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Oxidative damage of neurons is one of the key pathological markers of Alzheimer's disease (AD), which eventually leads to neuronal apoptosis and loss. Nuclear factor E2-related factor 2 (Nrf2) is a key regulator of antioxidant response and is considered to be an important therapeutic target for neurodegenerative diseases. In this study, the selenated derivative of antioxidant rutin (Se-Rutin) was synthesized with sodium selenate (Na2SeO3) as raw material by a simple electrostatic-compound in situ selenium reduction method. The effects of Se-Rutin on H2O2 induced oxidative damage in Pheochromocytoma PC12 cells were evaluated by cell viability, apoptosis, reactive oxygen species level and the expression of antioxidant response element (Nrf2). The results showed that H2O2 treatment significantly increased the level of apoptosis and reactive oxygen species, while the level of Nrf2 and HO-1 decreased. However, Se-Rutin significantly reduced H2O2 induced apoptosis and cytotoxicity, and increased the expression of Nrf2 and HO-1, both of which were better than that of pure rutin. Therefore, the activation of Nrf2/HO-1 signaling pathway may be the basis of Se-Rutin's anti-oxidative damage to AD.
Collapse
Affiliation(s)
- Mengyu Zhu
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Yang Zhang
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Chi Zhang
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Lei Chen
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Ying Kuang
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| |
Collapse
|
12
|
Lin NH, Goh A, Lin SH, Chuang KA, Chang CH, Li MH, Lu CH, Chen WY, Wei PH, Pan IH, Perng MD, Wen SF. Neuroprotective Effects of a Multi-Herbal Extract on Axonal and Synaptic Disruption in Vitro and Cognitive Impairment in Vivo. J Alzheimers Dis Rep 2023; 7:51-76. [PMID: 36777330 PMCID: PMC9912829 DOI: 10.3233/adr-220056] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
Background Alzheimer's disease (AD) is a multifactorial disorder characterized by cognitive decline. Current available therapeutics for AD have limited clinical benefit. Therefore, preventive therapies for interrupting the development of AD are critically needed. Molecules targeting multifunction to interact with various pathlogical components have been considered to improve the therapeutic efficiency of AD. In particular, herbal medicines with multiplicity of actions produce cognitive benefits on AD. Bugu-M is a multi-herbal extract composed of Ganoderma lucidum (Antler form), Nelumbo nucifera Gaertn., Ziziphus jujuba Mill., and Dimocarpus longan, with the ability of its various components to confer resilience to cognitive deficits. Objective To evaluate the potential of Bugu-M on amyloid-β (Aβ) toxicity and its in vitro mechanisms and on in vivo cognitive function. Methods We illustrated the effect of Bugu-M on Aβ25-35-evoked toxicity as well as its possible mechanisms to diminish the pathogenesis of AD in rat cortical neurons. For cognitive function studies, 2-month-old female 3×Tg-AD mice were administered 400 mg/kg Bugu-M for 30 days. Behavioral tests were performed to assess the efficacy of Bugu-M on cognitive impairment. Results In primary cortical neuronal cultures, Bugu-M mitigated Aβ-evoked toxicity by reducing cytoskeletal aberrations and axonal disruption, restoring presynaptic and postsynaptic protein expression, suppressing mitochondrial damage and apoptotic signaling, and reserving neurogenic and neurotrophic factors. Importantly, 30-day administration of Bugu-M effectively prevented development of cognitive impairment in 3-month-old female 3×Tg-AD mice. Conclusion Bugu-M might be beneficial in delaying the progression of AD, and thus warrants consideration for its preventive potential for AD.
Collapse
Affiliation(s)
- Ni-Hsuan Lin
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Angela Goh
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Shyh-Horng Lin
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Kai-An Chuang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chih-Hsuan Chang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Ming-Han Li
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chu-Hsun Lu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Wen-Yin Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Pei-Hsuan Wei
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - I-Hong Pan
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Ming-Der Perng
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan,
School of Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan,Correspondence to: Shu-Fang Wen, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, 321, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35743946; E-mail: and Ming-Der Perng, College of Life Sciences, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35742024; E-mail:
| | - Shu-Fang Wen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan,Correspondence to: Shu-Fang Wen, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, 321, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35743946; E-mail: and Ming-Der Perng, College of Life Sciences, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35742024; E-mail:
| |
Collapse
|
13
|
The Role of the NRF2 Pathway in Maintaining and Improving Cognitive Function. Biomedicines 2022; 10:biomedicines10082043. [PMID: 36009590 PMCID: PMC9405981 DOI: 10.3390/biomedicines10082043] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (NRF2) is a redox-sensitive transcription factor that binds to the antioxidant response element consensus sequence, decreasing reactive oxygen species and regulating the transcription of a wide array of genes, including antioxidant and detoxifying enzymes, regulating genes involved in mitochondrial function and biogenesis. Moreover, NRF2 has been shown to directly regulate the expression of anti-inflammatory mediators reducing the expression of pro-inflammatory cytokines. In recent years, attention has turned to the role NRF2 plays in the brain in different diseases such Alzheimer’s disease, Parkinson’s disease, Huntington’s disease and others. This review focused on the evidence, derived in vitro, in vivo and from clinical trials, supporting a role for NRF2 activation in maintaining and improving cognitive function and how its activation can be used to elicit neuroprotection and lead to cognitive enhancement. The review also brings a critical discussion concerning the possible prophylactic and/or therapeutic use of NRF2 activators in treating cognitive impairment-related conditions.
Collapse
|
14
|
Protective and anti-oxidative effects of curcumin and resveratrol on Aβ-oligomer-induced damage in the SH-SY5Y cell line. J Neurol Sci 2022; 441:120356. [DOI: 10.1016/j.jns.2022.120356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/27/2022] [Accepted: 07/24/2022] [Indexed: 11/18/2022]
|
15
|
Soejima-Kusunoki A, Okada K, Saito R, Watabe K. The Protective Effect of Edaravone on TDP-43 Plus Oxidative Stress-Induced Neurotoxicity in Neuronal Cells: Analysis of Its Neuroprotective Mechanisms Using RNA Sequencing. Pharmaceuticals (Basel) 2022; 15:ph15070842. [PMID: 35890141 PMCID: PMC9319738 DOI: 10.3390/ph15070842] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 02/06/2023] Open
Abstract
Edaravone is a free-radical scavenger drug that was recently approved for the treatment of amyotrophic lateral sclerosis (ALS), a neurodegenerative disease. A pathological hallmark of ALS is the accumulation of ubiquitinated or phosphorylated aggregates of the 43-kDa transactive response DNA binding protein (TDP-43) within the cytoplasm of motor neurons. This study revealed the efficacy of edaravone in preventing neuronal cell death in a TDP-43 proteinopathy model and analyzed the molecular changes associated with the neuroprotection. The viability of the neuronal cells expressing TDP-43 was reduced by oxidative stress, and edaravone (≥10 μmol/L) protected in a concentration-dependent manner against the neurotoxic insult. Differential gene expression analysis revealed changes among pathways related to nuclear erythroid 2-related-factor (Nrf2)-mediated oxidative stress response in cells expressing TDP-43. In edaravone-treated cells expressing TDP-43, significant changes in gene expression were also identified among Nrf2-oxidative response, unfolded protein response, and autophagy pathways. In addition, the expression of genes belonging to phosphatidylinositol metabolism pathways was modified. These findings suggest that the neuroprotective effect of edaravone involves the prevention of TDP-43 misfolding and enhanced clearance of pathological TDP-43 in TDP-43 proteinopathy.
Collapse
Affiliation(s)
- Aki Soejima-Kusunoki
- Research Unit of Neuroscience, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan
- Correspondence:
| | - Kinya Okada
- Discovery Technology Laboratories, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan; (K.O.); (R.S.)
| | - Ryuta Saito
- Discovery Technology Laboratories, Mitsubishi Tanabe Pharma Corporation, Yokohama 227-0033, Japan; (K.O.); (R.S.)
| | - Kazuhiko Watabe
- Faculty of Health Sciences, Kyorin University, Tokyo 181-8612, Japan;
| |
Collapse
|
16
|
Sharifyrad M, Gohari S, Fathi M, Danafar H, Hosseini MJ, Mostafavi H, Manjili HK. The efficacy and neuroprotective effects of edaravone-loaded mPEG-b-PLGA polymeric nanoparticles on human neuroblastoma SH-SY5Y cell line as in vitro model of ischemia. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
17
|
Arslanbaeva L, Bisaglia M. Activation of the Nrf2 Pathway as a Therapeutic Strategy for ALS Treatment. Molecules 2022; 27:1471. [PMID: 35268572 PMCID: PMC8911691 DOI: 10.3390/molecules27051471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 12/19/2022] Open
Abstract
Amyotrophic lateral sclerosis is a progressive and fatal disease that causes motoneurons degeneration and functional impairment of voluntary muscles, with limited and poorly efficient therapies. Alterations in the Nrf2-ARE pathway are associated with ALS pathology and result in aberrant oxidative stress, making the stimulation of the Nrf2-mediated antioxidant response a promising therapeutic strategy in ALS to reduce oxidative stress. In this review, we first introduce the involvement of the Nrf2 pathway in the pathogenesis of ALS and the role played by astrocytes in modulating such a protective pathway. We then describe the currently developed activators of Nrf2, used in both preclinical animal models and clinical studies, taking into consideration their potentialities as well as the possible limitations associated with their use.
Collapse
Affiliation(s)
| | - Marco Bisaglia
- Department of Biology, University of Padua, 35131 Padua, Italy
- Center Study for Neurodegeneration (CESNE), University of Padua, 35131 Padua, Italy
| |
Collapse
|
18
|
Cha SJ, Kim K. Effects of the Edaravone, a Drug Approved for the Treatment of Amyotrophic Lateral Sclerosis, on Mitochondrial Function and Neuroprotection. Antioxidants (Basel) 2022; 11:antiox11020195. [PMID: 35204078 PMCID: PMC8868074 DOI: 10.3390/antiox11020195] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 01/27/2023] Open
Abstract
Edaravone, the first known free radical scavenger, has demonstrated cellular protective properties in animals and humans. Owing to its antioxidant activity, edaravone modulates oxidative damage in various diseases, especially neurodegenerative diseases. In 2015, edaravone was approved in Japan to treat amyotrophic lateral sclerosis. The distinguishing pathogenic features of neurodegenerative diseases include high reactive oxygen species levels and mitochondrial dysfunction. However, the correlation between mitochondria and edaravone has not been elucidated. This review highlights recent studies on novel therapeutic perspectives of edaravone in terms of its effect on oxidative stress and mitochondrial function.
Collapse
Affiliation(s)
- Sun Joo Cha
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea;
| | - Kiyoung Kim
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea;
- Department of Medical Biotechnology, Soonchunhyang University, Asan 31538, Korea
- Correspondence: ; Tel.: +82-41-413-5024; Fax: +82-41-413-5006
| |
Collapse
|
19
|
Park HR, Yang EJ. Oxidative Stress as a Therapeutic Target in Amyotrophic Lateral Sclerosis: Opportunities and Limitations. Diagnostics (Basel) 2021; 11:diagnostics11091546. [PMID: 34573888 PMCID: PMC8465946 DOI: 10.3390/diagnostics11091546] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/14/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS), also known as motor neuron disease (MND) and Lou Gehrig’s disease, is characterized by a loss of the lower motor neurons in the spinal cord and the upper motor neurons in the cerebral cortex. Due to the complex and multifactorial nature of the various risk factors and mechanisms that are related to motor neuronal degeneration, the pathological mechanisms of ALS are not fully understood. Oxidative stress is one of the known causes of ALS pathogenesis. This has been observed in patients as well as in cellular and animal models, and is known to induce mitochondrial dysfunction and the loss of motor neurons. Numerous therapeutic agents have been developed to inhibit oxidative stress and neuroinflammation. In this review, we describe the role of oxidative stress in ALS pathogenesis, and discuss several anti-inflammatory and anti-oxidative agents as potential therapeutics for ALS. Although oxidative stress and antioxidant fields are meaningful approaches to delay disease progression and prolong the survival in ALS, it is necessary to investigate various animal models or humans with different subtypes of sporadic and familial ALS.
Collapse
|
20
|
Feng T, Yamashita T, Sasaki R, Tadokoro K, Matsumoto N, Hishikawa N, Abe K. Protective effects of edaravone on white matter pathology in a novel mouse model of Alzheimer's disease with chronic cerebral hypoperfusion. J Cereb Blood Flow Metab 2021; 41:1437-1448. [PMID: 33106078 PMCID: PMC8142121 DOI: 10.1177/0271678x20968927] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
White matter lesions (WMLs) caused by cerebral chronic hypoperfusion (CCH) may contribute to the pathophysiology of Alzheimer's disease (AD). However, the underlying mechanisms and therapeutic approaches have yet to be totally identified. In the present study, we investigated a potential therapeutic effect of the free radical scavenger edaravone (EDA) on WMLs in our previously reported novel mouse model of AD (APP23) plus CCH with motor and cognitive deficits. Relative to AD with CCH mice at 12 months (M) of age, EDA strongly improved CCH-induced WMLs in the corpus callosum of APP23 mice at 12 M by improving the disruption of white matter integrity, enhancing the proliferation of oligodendrocyte progenitor cells, attenuating endothelium/astrocyte unit dysfunction, and reducing neuroinflammation and oxidative stress. The present study demonstrates that the long-term administration of EDA may provide a promising therapeutic approach for WMLs in AD plus CCH disease with cognitive deficits.
Collapse
Affiliation(s)
- Tian Feng
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryo Sasaki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Koh Tadokoro
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Namiko Matsumoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Nozomi Hishikawa
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
21
|
Bhandari R, Kaur J, Kaur S, Kuhad A. The Nrf2 pathway in psychiatric disorders: pathophysiological role and potential targeting. Expert Opin Ther Targets 2021; 25:115-139. [PMID: 33557652 DOI: 10.1080/14728222.2021.1887141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: All psychiatric disorders exhibit excitotoxicity, mitochondrial dysfunction, inflammation, oxidative stress, and neural damage as their common characteristic. The endogenous nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway is implicated in the defense mechanism against oxidative stress and has a significant role in psychiatric disorders.Areas covered: We explore the role of Nrf2 pathway and its modulators in psychiatric disorders. The literature was searched utilizing various databases such as Embase, Medline, Web of Science, Pub-Med, and Google Scholar from 2010 to 2020. The search included research articles, clinical reports, systematic reviews, and meta-analyses.Expert opinion: Environmental factors and genetic predisposition can be a trigger for the development of psychiatric disorders. Nrf2 downregulates certain inflammatory pathways and upregulates various antioxidant enzymes to maintain a balance. However, its intricate balance with NF-Kβ (Nuclear factor kappa light chain enhancer of activated B cells) and its crosstalk with the transcription factor Nrf2 is critical in severe oxidative stress. Several Nrf2 modulators are now in clinical trials and can help reduce oxidative stress and neuroinflammation. There are immense potential opportunities for these modulators to become a novel therapeutic option.
Collapse
Affiliation(s)
- Ranjana Bhandari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Japneet Kaur
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Simerpreet Kaur
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| |
Collapse
|
22
|
Ismail H, Shakkour Z, Tabet M, Abdelhady S, Kobaisi A, Abedi R, Nasrallah L, Pintus G, Al-Dhaheri Y, Mondello S, El-Khoury R, Eid AH, Kobeissy F, Salameh J. Traumatic Brain Injury: Oxidative Stress and Novel Anti-Oxidants Such as Mitoquinone and Edaravone. Antioxidants (Basel) 2020; 9:antiox9100943. [PMID: 33019512 PMCID: PMC7601591 DOI: 10.3390/antiox9100943] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/04/2020] [Accepted: 09/22/2020] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) is a major health concern worldwide and is classified based on severity into mild, moderate, and severe. The mechanical injury in TBI leads to a metabolic and ionic imbalance, which eventually leads to excessive production of reactive oxygen species (ROS) and a state of oxidative stress. To date, no drug has been approved by the food and drug administration (FDA) for the treatment of TBI. Nevertheless, it is thought that targeting the pathology mechanisms would alleviate the consequences of TBI. For that purpose, antioxidants have been considered as treatment options in TBI and were shown to have a neuroprotective effect. In this review, we will discuss oxidative stress in TBI, the history of antioxidant utilization in the treatment of TBI, and we will focus on two novel antioxidants, mitoquinone (MitoQ) and edaravone. MitoQ can cross the blood brain barrier and cellular membranes to accumulate in the mitochondria and is thought to activate the Nrf2/ARE pathway leading to an increase in the expression of antioxidant enzymes. Edaravone is a free radical scavenger that leads to the mitigation of damage resulting from oxidative stress with a possible association to the activation of the Nrf2/ARE pathway as well.
Collapse
Affiliation(s)
- Helene Ismail
- Department of Neurology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.S.); (M.T.); (A.K.); (R.A.); (L.N.)
| | - Maha Tabet
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.S.); (M.T.); (A.K.); (R.A.); (L.N.)
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria 21544, Egypt;
| | - Abir Kobaisi
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.S.); (M.T.); (A.K.); (R.A.); (L.N.)
| | - Reem Abedi
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.S.); (M.T.); (A.K.); (R.A.); (L.N.)
| | - Leila Nasrallah
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.S.); (M.T.); (A.K.); (R.A.); (L.N.)
| | - Gianfranco Pintus
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah 27272, UAE;
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Yusra Al-Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 15551, UAE;
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98165 Messina, Italy;
| | - Riyad El-Khoury
- Department of Pathology and Laboratory Medicine, Neuromuscular Diagnostic Laboratory, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
| | - Ali H. Eid
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut 1107 2020, Lebanon
- Department of Biomedical Sciences, Qatar University, Doha 2713, Qatar
- Correspondence: (A.H.E.); (F.K.); (J.S.); Tel.: +961-1-350000 (ext. 4891) (A.H.E.); +961-1-350000 (ext. 4805) (F.K.); +961-1-350000 (ext. 7359) (J.S.)
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.S.); (M.T.); (A.K.); (R.A.); (L.N.)
- Correspondence: (A.H.E.); (F.K.); (J.S.); Tel.: +961-1-350000 (ext. 4891) (A.H.E.); +961-1-350000 (ext. 4805) (F.K.); +961-1-350000 (ext. 7359) (J.S.)
| | - Johnny Salameh
- Department of Neurology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
- Correspondence: (A.H.E.); (F.K.); (J.S.); Tel.: +961-1-350000 (ext. 4891) (A.H.E.); +961-1-350000 (ext. 4805) (F.K.); +961-1-350000 (ext. 7359) (J.S.)
| |
Collapse
|
23
|
Boiangiu RS, Mihasan M, Gorgan DL, Stache BA, Petre BA, Hritcu L. Cotinine and 6-Hydroxy-L-Nicotine Reverses Memory Deficits and Reduces Oxidative Stress in Aβ 25-35-Induced Rat Model of Alzheimer's Disease. Antioxidants (Basel) 2020; 9:E768. [PMID: 32824768 PMCID: PMC7465470 DOI: 10.3390/antiox9080768] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/08/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
The nicotinic derivatives, cotinine (COT), and 6-hydroxy-L-nicotine (6HLN), showed promising cognitive-improving effects without exhibiting the nicotine's side-effects. Here, we investigated the impact of COT and 6HLN on memory impairment and the oxidative stress in the Aβ25-35-induced rat model of Alzheimer's disease (AD). COT and 6HLN were chronically administered to Aβ25-35-treated rats, and their memory performances were assessed using in vivo tasks (Y-maze, novel object recognition, and radial arm maze). By using in silico tools, we attempted to associate the behavioral outcomes with the calculated binding potential of these nicotinic compounds in the allosteric sites of α7 and α4β2 subtypes of the nicotinic acetylcholine receptors (nAChRs). The oxidative status and acetylcholinesterase (AChE) activity were determined from the hippocampal tissues. RT-qPCR assessed bdnf, arc, and il-1β mRNA levels. Our data revealed that COT and 6HLN could bind to α7 and α4β2 nAChRs with similar or even higher affinity than nicotine. Consequently, the treatment exhibited a pro-cognitive, antioxidant, and anti-AChE profile in the Aβ25-35-induced rat model of AD. Finally, RT-qPCR analysis revealed that COT and 6HLN positively modulated the bdnf, arc, and il-1β genes expression. Therefore, these nicotinic derivatives that act on the cholinergic system might represent a promising choice to ameliorate AD conditions.
Collapse
Affiliation(s)
- Razvan Stefan Boiangiu
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 700506 Iasi, Romania; (R.S.B.); (M.M.); (D.L.G.); (B.A.S.)
| | - Marius Mihasan
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 700506 Iasi, Romania; (R.S.B.); (M.M.); (D.L.G.); (B.A.S.)
| | - Dragos Lucian Gorgan
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 700506 Iasi, Romania; (R.S.B.); (M.M.); (D.L.G.); (B.A.S.)
| | - Bogdan Alexandru Stache
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 700506 Iasi, Romania; (R.S.B.); (M.M.); (D.L.G.); (B.A.S.)
- Center for Fundamental Research and Experimental Development in Translation Medicine—TRANSCEND, Regional Institute of Oncology, 700483 Iasi, Romania;
| | - Brindusa Alina Petre
- Center for Fundamental Research and Experimental Development in Translation Medicine—TRANSCEND, Regional Institute of Oncology, 700483 Iasi, Romania;
- Department of Chemistry, Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 700506 Iasi, Romania
| | - Lucian Hritcu
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 700506 Iasi, Romania; (R.S.B.); (M.M.); (D.L.G.); (B.A.S.)
| |
Collapse
|
24
|
1,25(OH) 2D 3 Alleviates Aβ(25-35)-Induced Tau Hyperphosphorylation, Excessive Reactive Oxygen Species, and Apoptosis Through Interplay with Glial Cell Line-Derived Neurotrophic Factor Signaling in SH-SY5Y Cells. Int J Mol Sci 2020; 21:ijms21124215. [PMID: 32545801 PMCID: PMC7352552 DOI: 10.3390/ijms21124215] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 01/14/2023] Open
Abstract
Amyloid beta (Aβ) accumulation in the brain is one of the major pathological features of Alzheimer's disease. The active form of vitamin D (1,25(OH)2D3), which acts via its nuclear hormone receptor, vitamin D receptor (VDR), has been implicated in the treatment of Aβ pathology, and is thus considered as a neuroprotective agent. However, its underlying molecular mechanisms of action are not yet fully understood. Here, we aim to investigate whether the molecular mechanisms of 1,25(OH)2D3 in ameliorating Aβ toxicity involve an interplay of glial cell line-derived neurotrophic factor (GDNF)-signaling in SH-SY5Y cells. Cells were treated with Aβ(25-35) as the source of toxicity, followed by the addition of 1,25(OH)2D3 with or without the GDNF inhibitor, heparinase III. The results show that 1,25(OH)2D3 modulated Aβ-induced reactive oxygen species, apoptosis, and tau protein hyperphosphorylation in SH-SY5Y cells. Additionally, 1,25(OH)2D3 restored the decreasing GDNF and the inhibited phosphorylation of the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt)/glycogen synthase kinase-3β (GSK-3β) protein expressions. In the presence of heparinase III, these damaging effects evoked by Aβ were not abolished by 1,25(OH)2D3. It appears 1,25(OH)2D3 is beneficial for the alleviation of Aβ neurotoxicity, and it might elicit its neuroprotection against Aβ neurotoxicity through an interplay with GDNF-signaling.
Collapse
|
25
|
Potential use of edaravone to reduce specific side effects of chemo-, radio- and immuno-therapy of cancers. Int Immunopharmacol 2019; 77:105967. [DOI: 10.1016/j.intimp.2019.105967] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/30/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023]
|