1
|
Altahrawi AY, James AW, Shah ZA. The Role of Oxidative Stress and Inflammation in the Pathogenesis and Treatment of Vascular Dementia. Cells 2025; 14:609. [PMID: 40277934 PMCID: PMC12026122 DOI: 10.3390/cells14080609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
Vascular dementia (VaD) is a heterogeneous group of brain disorders caused by cerebrovascular pathologies and the second most common cause of dementia, accounting for over 20% of cases and posing an important global health concern. VaD can be caused by cerebral infarction or injury in critical brain regions, including the speech area of the dominant hemisphere or arcuate fasciculus of the dominant hemisphere, leading to notable cognitive impairment. Although the exact causes of dementia remain multifactorial and complex, oxidative stress (reactive oxygen species), neuroinflammation (TNFα, IL-6, and IL-1β), and inflammasomes are considered central mechanisms in its pathology. These conditions contribute to neuronal damage, synaptic dysfunction, and cognitive decline. Thus, antioxidants and anti-inflammatory agents have emerged as potential therapeutic targets in dementia. Recent studies emphasize that cerebrovascular disease plays a dual role: first, as a primary cause of cognitive impairment and then as a contributor to the manifestation of dementia driven by other factors, such as Alzheimer's disease and other neurodegenerative conditions. This comprehensive review of VaD focuses on molecular mechanisms and their consequences. We provided up-to-date knowledge about epidemiology, pathophysiological mechanisms, and current therapeutic approaches for VaD.
Collapse
Affiliation(s)
- Aseel Y. Altahrawi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Antonisamy William James
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA;
| | - Zahoor A. Shah
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA;
| |
Collapse
|
2
|
Barati A, Moghimi S, Taghavi Zanjani K, Rohani M, Sohrabi Hesar M, Arfaie A, Ghezelche Khamsiyan M, Mahmoudi J, Sadigh-Eteghad S. Acute Administration of Edaravone Improves Cognitive Impairment in a Mouse Model of mPFC Ischemia: Crosstalk Between Necroptosis, Neuroinflammation, and Antioxidant Defense. Mol Neurobiol 2025; 62:4420-4434. [PMID: 39448519 DOI: 10.1007/s12035-024-04541-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024]
Abstract
Edaravone (Eda), a well-known free radical scavenger, has been reported as a possible therapeutic agent for ischemic stroke patients' recovery. This study aimed to investigate the effects of time-dependent treatment with Eda on medial prefrontal cortex (mPFC) ischemia. Mice were randomly allocated into six groups: control, sham, normal saline, Eda-I, Eda-II, and Eda-III. After induction of a photothrombotic ischemia in the mPFC region, Eda-I, Eda-II, and Eda-III groups received 3 mg/kg Eda intraperitoneally at the times of 0, 2, and 6 h post-surgery. After 1 day of recovery, the mice underwent behavioral tests (open field, novel object recognition, and T-maze). Next, necroptosis, NOD-like receptor protein 3 (NLRP3), and nuclear factor erythroid 2-related factor 2 (Nrf2) pathway-related protein levels were measured in the lesioned area using western blot analysis. For double confirmation, IL-1β and IL-18 were also assessed by immunofluorescence in the area. Further, histological evaluations were performed to measure tissue damage. The results showed that mPFC ischemia impaired recognition and spatial working memory without affecting locomotor activity, while immediate Eda administration improved cognitive impairments. Furthermore, acute Eda treatment reduced RIP1, RIP3, and MLKL levels, inhibited NLRP3 inflammasome proteins (NLRP3, ASC, and Cas1), decreased IL-1β and IL-18, upregulated Nrf2 and its targets (NQO-1 and HO-1), and diminished tissue damage. Our results highlighted the effects of acute administration of Eda post-stroke on improving cognitive impairments by suppressing necroptosis and NLRP3 inflammasome pathways and activating the Nrf2 antioxidant defense mechanism.
Collapse
Affiliation(s)
- Alireza Barati
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sadegh Moghimi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kiana Taghavi Zanjani
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Mojde Rohani
- Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, Iran
| | - Mehri Sohrabi Hesar
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Arian Arfaie
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | | | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Kimura S, Iwata M, Takase H, Lo EH, Arai K. Oxidative stress and chronic cerebral hypoperfusion: An overview from preclinical rodent models. J Cereb Blood Flow Metab 2025; 45:381-395. [PMID: 39663901 PMCID: PMC11635795 DOI: 10.1177/0271678x241305899] [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] [Received: 06/24/2024] [Revised: 10/12/2024] [Accepted: 11/19/2024] [Indexed: 12/13/2024]
Abstract
Chronic cerebral hypoperfusion (CCH) is an important clinical condition characterized by a prolonged reduction in cerebral blood flow that contributes to several neurodegenerative diseases, including vascular dementia and Alzheimer's disease. A number of rodent models of CCH have been developed that mimic the human pathological conditions of reduced cerebral perfusion. These models have been instrumental in elucidating the molecular and cellular mechanisms involved in CCH-induced brain damage. Oxidative stress is induced by perturbations in cellular pathways caused by CCH, including mitochondrial dysfunction, ion pump dysfunction, and adenosine triphosphate (ATP) depletion. The deleterious stress leads to the accumulation of reactive oxygen species (ROS) and exacerbates damage to neuronal structures, significantly impairing cognitive function. Among the various therapeutic strategies being evaluated, edaravone, a potent antioxidant, is emerging as a promising drug due to its neuroprotective properties against oxidative stress. Initially approved for use in ischemic stroke, research using rodent CCH models has shown that edaravone has significant efficacy in scavenging free radicals and ameliorating oxidative stress-induced neuronal damage under CCH conditions. This mini-review summarizes the current literature on the rodent models of CCH and then discusses the therapeutic potential of edaravone to reduce neuronal and vascular damage caused by CCH-induced oxidative stress.
Collapse
Affiliation(s)
- Shintaro Kimura
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Life Science Research Center, Gifu University, Gifu, Japan
| | - Maho Iwata
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Developmental Neuroscience, Tohoku University School of Medicine, Sendai, Japan
| | - Hajime Takase
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Dang C, Wang Q, Zhuang Y, Li Q, Feng L, Xiong Y, Lu Y. Pharmacological treatments for vascular dementia: a systematic review and Bayesian network meta-analysis. Front Pharmacol 2024; 15:1451032. [PMID: 39239652 PMCID: PMC11374729 DOI: 10.3389/fphar.2024.1451032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/26/2024] [Indexed: 09/07/2024] Open
Abstract
Background Vascular dementia (VaD) is one of the most prevalent, burdensome, and costly forms of dementia. Pharmacological treatment is often the first-line choice for clinicians; however, there is a paucity of comparative information regarding the multiple available drug options. Methods and Analysis A systematic review and network meta-analysis were conducted on randomized trials involving adult patients with VaD, sourced from PubMed, the Cochrane Library, EMBASE, Web of Science, OPENGREY, ClinicalTrials.gov, Wanfang Data, and CNKI. The primary outcomes included changes in Mini-Mental State Examination (MMSE) scores, activities of daily living (ADL) scores, and the incidence of adverse reactions. Efficacy and safety of intervention strategies were comprehensively analyzed using forest plots, cumulative ranking probability curves (SUCRA), and funnel plots, all generated with R software. Results A total of 194 RCTs comparing 21 different anti-VaD drugs with placebos or no treatment were analysed. Regarding MMSE scores, the five most effective drugs were Butylphthalide, Huperzine A, Edaravone, Rivastigmine, and Memantine. For ADL scores, the top five drugs in efficacy were Huperzine A, Butylphthalide, Tianzhi granule, Nicergoline, and Idebenone. In terms of the incidence of adverse drug reactions, Co-dergocrine Mesylate, Tongxinluo capsule, Butylphthalide, Piracetam, and Oxiracetam demonstrated favourable safety profiles. Conclusion This study enhances the understanding of the relative benefits and risks associated with various VaD treatments, providing a valuable reference for clinical decision-making. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/, identifier registration number.
Collapse
Affiliation(s)
- Chun Dang
- Department of Periodical Press, West China Hospital, Sichuan University, Chengdu, China
| | - Qinxuan Wang
- West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yijia Zhuang
- West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Qian Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Feng
- Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Xiong
- Department of Periodical Press, West China Hospital, Sichuan University, Chengdu, China
| | - Yaoheng Lu
- Department of General Surgery, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Chengdu, China
| |
Collapse
|
5
|
Wei N, Zhang LM, Xu JJ, Li SL, Xue R, Ma SL, Li C, Sun MM, Chen KS. Astaxanthin Rescues Memory Impairments in Rats with Vascular Dementia by Protecting Against Neuronal Death in the Hippocampus. Neuromolecular Med 2024; 26:29. [PMID: 39014255 DOI: 10.1007/s12017-024-08796-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
Vascular dementia (VaD) is a cognitive disorder characterized by a decline in cognitive function resulting from cerebrovascular disease. The hippocampus is particularly susceptible to ischemic insults, leading to memory deficits in VaD. Astaxanthin (AST) has shown potential therapeutic effects in neurodegenerative diseases. However, the mechanisms underlying its protective effects in VaD and against hippocampal neuronal death remain unclear. In this study, We used the bilateral common carotid artery occlusion (BCCAO) method to establish a chronic cerebral hypoperfusion (CCH) rat model of VaD and administered a gastric infusion of AST at 25 mg/kg per day for 4 weeks to explore its therapeutic effects. Memory impairments were assessed using Y-maze and Morris water maze tests. We also performed biochemical analyses to evaluate levels of hippocampal neuronal death and apoptosis-related proteins, as well as the impact of astaxanthin on the PI3K/Akt/mTOR pathway and oxidative stress. Our results demonstrated that AST significantly rescued memory impairments in VaD rats. Furthermore, astaxanthin treatment protected against hippocampal neuronal death and attenuated apoptosis. We also observed that AST modulated the PI3K/Akt/mTOR pathway, suggesting its involvement in promoting neuronal survival and synaptic plasticity. Additionally, AST exhibited antioxidant properties, mitigating oxidative stress in the hippocampus. These findings provide valuable insights into the potential therapeutic effects of AST in VaD. By elucidating the mechanisms underlying the actions of AST, this study highlights the importance of protecting hippocampal neurons and suggests potential targets for intervention in VaD. There are still some unanswered questions include long-term effects and optimal dosage of the use in human. Further research is warranted to fully understand the therapeutic potential of AST and its application in the clinical treatment of VaD.
Collapse
Affiliation(s)
- Na Wei
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China.
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China.
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China.
| | - Luo-Man Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China
| | - Jing-Jing Xu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China
| | - Sheng-Lei Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
| | - Sheng-Li Ma
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
| | - Cai Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, Zhengzhou, 450002, People's Republic of China
| | - Miao-Miao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China
| | - Kui-Sheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Henan Key Laboratory of Tumor Pathology, No.1 Jian She Dong Avenue, Zhengzhou, 450002, People's Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, No.100 Ke Xue Avenue, Zhengzhou, 450002, People's Republic of China
| |
Collapse
|
6
|
Fessel J. Cure of Alzheimer's Dementia in Many Patients by Using Intranasal Insulin to Augment an Inadequate Counter-Reaction, Edaravone to Scavenge ROS, and 1 or 2 Other Drugs to Address Affected Brain Cells. J Clin Med 2023; 12:jcm12093151. [PMID: 37176592 PMCID: PMC10178959 DOI: 10.3390/jcm12093151] [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: 03/09/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
The goal of treatment for Alzheimer's dementia (AD) is the restoration of normal cognition. No drug regimen has ever achieved this. This article suggests that curing AD may be achieved by combination therapy as follows. First, with intranasal insulin to augment the body's natural counter-reaction to the changes in brain cell-types that produced the dementia. Second, with edaravone to decrease free radicals, which are increased and causal in AD. Third, as described elsewhere, with one or two drugs from among pioglitazone, fluoxetine, and lithium, which address the brain cell-types whose changed functions cause the dementia. Insulin restores cerebral glucose, which is the main nutrient for brain neurons whose depletion is responsible for the dementia; and edaravone decreases ROS, which are intrinsic causes of neuropathology in AD. This combination of drugs is a potential cure for many patients with AD, and should be tested in a clinical trial.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California San Francisco, 2069 Filbert Street, San Francisco, CA 94123, USA
| |
Collapse
|
7
|
Weerasinghe-Mudiyanselage PDE, Ang MJ, Kang S, Kim JS, Moon C. Structural Plasticity of the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:3349. [PMID: 35328770 PMCID: PMC8955928 DOI: 10.3390/ijms23063349] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/10/2022] Open
Abstract
Neuroplasticity is the capacity of neural networks in the brain to alter through development and rearrangement. It can be classified as structural and functional plasticity. The hippocampus is more susceptible to neuroplasticity as compared to other brain regions. Structural modifications in the hippocampus underpin several neurodegenerative diseases that exhibit cognitive and emotional dysregulation. This article reviews the findings of several preclinical and clinical studies about the role of structural plasticity in the hippocampus in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. In this study, literature was surveyed using Google Scholar, PubMed, Web of Science, and Scopus, to review the mechanisms that underlie the alterations in the structural plasticity of the hippocampus in neurodegenerative diseases. This review summarizes the role of structural plasticity in the hippocampus for the etiopathogenesis of neurodegenerative diseases and identifies the current focus and gaps in knowledge about hippocampal dysfunctions. Ultimately, this information will be useful to propel future mechanistic and therapeutic research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Poornima D. E. Weerasinghe-Mudiyanselage
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Mary Jasmin Ang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
- College of Veterinary Medicine, University of the Philippines Los Baños, Los Baños 4031, Philippines
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| |
Collapse
|
8
|
Alqudah MA, Al-Nosairy A, Alzoubi KH, Kahbour OF, Alazzam SI. Edaravone prevents memory impairment in diabetic rats: Role of oxidative stress. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
9
|
Meftahi GH, Bayat M, Zarifkar AH, Akbari S, Borhani Haghighi A, Naseh M, Yousefi Nejad A, Haghani M. Treatment with edaravone improves the structure and functional changes in the hippocampus after chronic cerebral hypoperfusion in rat. Brain Res Bull 2021; 174:122-130. [PMID: 34116172 DOI: 10.1016/j.brainresbull.2021.06.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 02/08/2023]
Abstract
This study aimed to find out cellular and electrophysiological effects of the edaravone (EDR) administration following induction of vascular dementia (VaD) via bilateral-carotid vessel occlusion (2VO). The rats were randomly divided into control, sham, 2VO + V (vehicle), and 2VO + EDR groups. EDR was administered once a day from day 0-28 after surgery. The passive-avoidance, Morris water-maze, and open-field tests were used for evaluation of memory, locomotor, and anxiety. The field-potential recording was used for assessment of electrophysiological properties of the hippocampus; and after sacrificing, the cerebral hemispheres were removed for stereological study and evaluation of MDA levels. The long-term potentiation (LTP), paired-pulse ratio (PPR), and input-output (I/O) curves were evaluated as indexes for long-term and short-term synaptic plasticity, and basal-synaptic transmission (BST), respectively. The 2VO led to increases in MDA level with considerable neuronal loss and decreases in the volume of the hippocampus, along with a reduction in the BST and LTP induction which was associated with a decrement in PPR and ultimate loss in memory with higher anxiety behavior. However, administration of EDR caused a decline in MDA and prevented the neural loss and volume of the hippocampus, rescued BST and LTP depression, improved memory and anxiety without any effects on PPR. Therefore, most likely through the improvement of MDA level, and the hippocampal cell number and volume, EDR leads to recovery of synaptic plasticity and behavioral performance. Because of the LTP rescue, without recovery of PPR, it is likely that the EDR improved LTP through the post-synaptic neurons.
Collapse
Affiliation(s)
- Gholam Hossein Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mahnaz Bayat
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Hossein Zarifkar
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Somaye Akbari
- Department of Physiology, the Medical School, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | | - Maryam Naseh
- Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amirhossein Yousefi Nejad
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, Islamic Azad University of Kazeroon, Shiraz, Iran.
| | - Masoud Haghani
- Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
10
|
Wei C, Xu X, Zhu H, Zhang X, Gao Z. Promotive role of microRNA‑150 in hippocampal neurons apoptosis in vascular dementia model rats. Mol Med Rep 2021; 23:257. [PMID: 33576461 PMCID: PMC7893740 DOI: 10.3892/mmr.2021.11896] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/21/2020] [Indexed: 01/22/2023] Open
Abstract
Cognitive impairment is one of the primary features of vascular dementia (VD). However, the specific mechanism underlying the regulation of cognition function in VD is not completely understood. The present study aimed to explore the effects of microRNA (miR)‑150 on VD. To determine the effects of miR‑150 on cognitive function and hippocampal neurons in VD model rats, rats were subjected to intracerebroventricular injections of miR‑150 antagomiR. The Morris water maze test results demonstrated that spatial learning ability was impaired in VD model rats compared with control rats. Moreover, compared with antagomiR negative control (NC), miR‑150 antagomiR alleviated cognitive impairment and enhanced memory ability in VD model rats. The triphenyltetrazolium chloride, Nissl staining and immunohistochemistry results further demonstrated that miR‑150 knockdown improved the activity of hippocampal neurons in VD model rats compared with the antagomiR NC group. To validate the role of miR‑150 in neurons in vitro, the PC12 cell line was used. The flow cytometry and Hoechst 33342/PI double staining results indicated that miR‑150 overexpression significantly increased cell apoptosis compared with the mimic NC group. Moreover, the dual‑luciferase reporter gene assay results indicated that miR‑150 targeted HOXA1 and negatively regulated HOXA1 expression. Therefore, the present study indicated that miR‑150 knockdown ameliorated VD symptoms by upregulating HOXA1 expression in vivo and in vitro.
Collapse
Affiliation(s)
- Chengqun Wei
- Department of General Practice, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Xiuzhi Xu
- Department of General Practice, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Hongyan Zhu
- Department of General Practice, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Xiuyan Zhang
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Zhan Gao
- Department of General Practice, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
11
|
Kuang H, Zhou ZF, Zhu YG, Wan ZK, Yang MW, Hong FF, Yang SL. Pharmacological Treatment of Vascular Dementia: A Molecular Mechanism Perspective. Aging Dis 2021; 12:308-326. [PMID: 33532143 PMCID: PMC7801279 DOI: 10.14336/ad.2020.0427] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/27/2020] [Indexed: 11/01/2022] Open
Abstract
Vascular dementia (VaD) is a neurodegenerative disease, with cognitive dysfunction attributable to cerebrovascular factors. At present, it is the second most frequently occurring type of dementia in older adults (after Alzheimer's disease). The underlying etiology of VaD has not been completely elucidated, which limits its management. Currently, there are no approved standard treatments for VaD. The drugs used in VaD are only suitable for symptomatic treatment and cannot prevent or reduce the occurrence and progression of VaD. This review summarizes the current status of pharmacological treatment for VaD, from the perspective of the molecular mechanisms specified in various pathogenic hypotheses, including oxidative stress, the central cholinergic system, neuroinflammation, neuronal apoptosis, and synaptic plasticity. As VaD is a chronic cerebrovascular disease with multifactorial etiology, combined therapy, targeting multiple pathophysiological factors, may be the future trend in VaD.
Collapse
Affiliation(s)
- Huang Kuang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, China.
| | - Zhi-Feng Zhou
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, China.
| | - Yu-Ge Zhu
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, China.
| | - Zhi-Kai Wan
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, China.
| | - Mei-Wen Yang
- Department of Nurse, Nanchang University Hospital, Nanchang 330006, Jiangxi, China.
| | - Fen-Fang Hong
- Department of Experimental Teaching Center, Nanchang University, Nanchang, China.
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, China.
- Department of Experimental Teaching Center, Nanchang University, Nanchang, China.
| |
Collapse
|
12
|
Yi XX, Li JY, Tang ZZ, Jiang S, Liu YH, Deng JG, Gao CH. Marinoid J, a phenylglycoside from Avicennia marina fruit, ameliorates cognitive impairment in rat vascular dementia: a quantitative iTRAQ proteomic study. PHARMACEUTICAL BIOLOGY 2020; 58:1211-1220. [PMID: 33280468 PMCID: PMC7723022 DOI: 10.1080/13880209.2020.1837187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 04/11/2020] [Accepted: 10/09/2020] [Indexed: 06/12/2023]
Abstract
CONTEXT Fruit of Avicennia marina (Forsk.) Vierh. (Acanthaceae) is used as a Chinese herb. Studies have found that it contains marinoid J, a novel phenylethanoid glycoside (PG) compound, but its neuroprotective functions are largely unknown. OBJECTIVE This study evaluated the effects of marinoid J on vascular dementia (VD) and determined its potential mechanisms of action. MATERIALS AND METHODS The VD model was established by the ligation of the bilateral common carotid artery in Sprague-Dawley rats, who received daily intragastrically administration of saline, marinoid J (125 or 500 mg/kg body weight/d), or oxiracetam (250 mg/kg body weight/d) for 14 days (20 rats in each group). The Morris water maze (MWM) was used to evaluate cognitive performance. The hippocampus was subjected to histological and proteomic analyses. RESULTS Marinoid J shortened the escape latency of VD rats (31.07 ± 3.74 s, p < 0.05). It also decreased malondialdehyde (MDA) (27.53%) and nitric oxide (NO) (20.41%) while increasing superoxide dismutase (SOD) (11.26%) and glutathione peroxidase (GSH-Px) (20.38%) content in hippocampus tissues. Proteomic analysis revealed 45 differentially expressed proteins (DEPs) in marinoid J-treated VD rats, which included angiotensin-converting enzyme (ACE), keratin 18 (KRT18), cluster of differentiation 34 (CD34), and synaptotagmin II (SYT2). CONCLUSIONS Marinoid J played a role in protecting hippocampal neurons by regulating a set of proteins that influence oxidative stress and apoptosis, this effect may thereby alleviate the symptoms of VD rats. Thus, pharmacological manipulation of marinoid J may offer a novel opportunity for VD treatment.
Collapse
Affiliation(s)
- Xiang-xi Yi
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Guangxi, China
- Faculty of Pharmacy, Guangxi University of Chinese Medicine, Guangxi, China
| | - Jia-yi Li
- Faculty of Pharmacy, Guangxi University of Chinese Medicine, Guangxi, China
| | - Zhen-zhou Tang
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Guangxi, China
| | - Shu Jiang
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Guangxi, China
| | - Yong-hong Liu
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Guangxi, China
| | - Jia-gang Deng
- Faculty of Pharmacy, Guangxi University of Chinese Medicine, Guangxi, China
| | - Cheng-hai Gao
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Guangxi, China
| |
Collapse
|
13
|
Alzoubi KH, Aburashed ZO, Mayyas F. Edaravone protects from memory impairment induced by chronic L-methionine administration. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:1221-1228. [PMID: 31989235 DOI: 10.1007/s00210-020-01827-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 01/17/2020] [Indexed: 02/06/2023]
Abstract
Hyperhomocysteinemia is a well-known cause of cognitive impairment and neurodegeneration. Increased oxidative stress in the brain has a major possible role in hyperhomocysteinemia-induced pathogenesis. Edaravone is a potent free radical scavenger that has a neuroprotective effect against memory impairment in several experimental models. The current study investigated the possible protective effect of edaravone in L-methionine-induced vascular dementia in a rat model. L-methionine was given (1.7 mg/kg/day) through oral gavage, while edaravone was given (6 mg/kg/day) intraperitoneally. The administration of methionine and edaravone started concomitantly and continued for a total of 9 weeks. Spatial learning and memory were assessed using the radial arm water maze (RAWM). Changes in the oxidative stress-related biomarkers in the hippocampus were assessed using enzymatic assays. Chronic L-methionine administration resulted in short-term and long-term memory impairment, whereas edaravone prevented such effect. Furthermore, edaravone ameliorated L-methionine induced decrease in the activity of the antioxidant enzymes catalase and glutathione peroxidase as well as the ratio of reduced glutathione to oxidized glutathione (GSH/GSSG ratio). Edaravone also prevented increase in the oxidized glutathione (GSSG) secondary to chronic L-methionine administration. In conclusion, the current study suggests that memory impairment and oxidative stress secondary to chronic L-methionine administration can be prevented by edaravone, probably via enhancing antioxidant mechanisms in the hippocampus.
Collapse
Affiliation(s)
- Karem H Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Zainah O Aburashed
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Fadia Mayyas
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
14
|
Singh S, Kumar A. Protective Effect of Edaravone on Cyclophosphamide Induced Oxidative Stress and Neurotoxicity in Rats. Curr Drug Saf 2020; 14:209-216. [PMID: 31057112 PMCID: PMC6864589 DOI: 10.2174/1574886314666190506100717] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/11/2019] [Accepted: 04/24/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Cyclophosphamide (CPA) is the most widely prescribed cancer chemotherapeutic agent which shows serious neurotoxic side effect. Generation of reactive oxygen species at the cellular level is the basic mechanism of cyclophosphamide induced neurotoxicity. Edaravone is the synthetic drug used for brain stroke and has potent antioxidant property. OBJECTIVE This study aimed to investigate the effect of edaravone on neurobehavioral and neuropathological alteration induced by cyclophosphamide in male rats. METHODS Twenty eight Sprague-Dawley rats were equally divided into four groups of seven rats in each. The control group received saline, and other groups were given CPA intraperitoneally (100 mg/kg), CPA (100 mg/kg) intraperitoneally + Edaravone (10 mg/kg) orally, or Edaravone (10 mg/kg) orally for one month. RESULTS Our data showed that CPA significantly elevated brain AChE activity in the hippocampal region. A decrease in the total antioxidant capacity and a reduction in the CAT, SOD, and GPX activity occurred in the brains of the rats exposed to CPA. CPA-treated rats showed a significant impairment in long-termmemory and motor coordination. These results were supported by histopathological observations of the brain. Results revealed that administration of edaravone reversed AChE activity alternation and ameliorated behavioral and histopathological changes induced by CPA. CONCLUSION This study suggests that co-administration of edaravone with cyclophosphamide may be a useful intriguing therapeutic approach to overcome cyclophosphamide induced neurotoxicity.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India.,Truba Institute of Pharmacy, Bhopal, MP, India
| | | |
Collapse
|
15
|
Abstract
Post-traumatic stress disorder (PTSD) is a mental health problem that develops in a proportion of individuals after experiencing a potential life-threatening traumatic stress event. Edaravone is a free radical scavenger, with a neuroprotective effect against cognitive impairment in several animal models. In the present study, the protective effect of edaravone on PTSD-induced memory impairment was investigated. Single prolonged stress was used as an animal model of PTSD, comprising 2 h of restrain, 20-min forced swimming, 15-min rest, and 1-2-min diethyl ether exposure. Concurrently, edaravone was given at a dose of 6 mg/kg/day, intraperitoneally, for 21 days. The radial arm water maze was used to assess learning and memory. Antioxidant biomarkers were measured in hippocampus tissues. Chronic administration of edaravone prevented impairment of short-term and long-term memory. Edaravone also prevented the stress-induced decrease in the ratio of reduced glutathione/oxidized glutathione and the activities of glutathione peroxidase and catalase enzymes in the hippocampus, as well as increases in the levels of oxidized glutathione and thiobarbituric acid reactive substances. In conclusion, edaravone ameliorated oxidative stress and cognitive impairment associated with a PTSD model, probably by supporting antioxidant mechanism in the hippocampus.
Collapse
|
16
|
Nanotherapy for Alzheimer's disease and vascular dementia: Targeting senile endothelium. Adv Colloid Interface Sci 2018; 251:44-54. [PMID: 29274774 DOI: 10.1016/j.cis.2017.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 12/12/2022]
Abstract
Due to the complexity of Alzheimer's disease, multiple cellular types need to be targeted simultaneously in order for a given therapy to demonstrate any major effectiveness. Ultrasound-sensitive coated microbubbles (in a targeted lipid nanoemulsion) are available. Versatile small molecule drug(s) targeting multiple pathways of Alzheimer's disease pathogenesis are known. By incorporating such drug(s) into the targeted "lipid-coated microbubble" [LCM]/"nanoparticle-derived" [ND] (or LCM/ND) nanoemulsion type, one obtains a multitasking combination therapeutic for translational medicine. This multitasking therapeutic targets cell-surface scavenger receptors (mainly class B type I), or SR-BI, making possible for various Alzheimer's-related cell types to be simultaneously searched out for localized drug treatment in vivo. Besides targeting cell-surface SR-BI, the proposed LCM/ND-nanoemulsion combination therapeutic(s) include a characteristic lipid-coated microbubble [LCM] subpopulation (i.e., a stable LCM suspension); such film-stabilized microbubbles are well known to substantially reduce the acoustic power levels needed for accomplishing temporary noninvasive (transcranial) ultrasound treatment, or sonoporation, if additionally desired for the Alzheimer's patient.
Collapse
|