1
|
Moradi Khankani A, Hossein Meftahi G. Pretreatment with 4-methylumbilliferon improves anxiety-like behaviors and memory impairment in stressed rats via modulation of neuronal cell death and oxidative stress. Brain Res 2024; 1844:149196. [PMID: 39181223 DOI: 10.1016/j.brainres.2024.149196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
This work was done to investigate the ameliorating impact of 4-methylumbilliferon (4-MU) on spatial learning and memory dysfunction and restraint stress (STR)-induced anxiety-like behaviors in male Wistar rats and the underlying mechanisms. Thirty-two animals were assigned into 4 cohorts: control, 4-MU, STR, and STR+4-MU. Animals were exposed to STR for 4 h per day for 14 consecutive days or kept in normal conditions (healthy animals without exposure to stress). 4-MU (25 mg/kg) was intraperitoneally administered once daily to STR rats before restraint stress for 14 consecutive days. The behavioral tests were performed through Morris water maze tests and elevated-plus maze to examine learning/memory function, and anxiety levels, respectively. The levels of the antioxidant defense biomarkers (GPX, SOD) and MDA as an oxidant molecule in the brain tissues were measured using commercial ELISA kits. Neuronal loss or density of neurons was evaluated using Nissl staining. STR exposure could cause significant alterations in the levels of the antioxidant defense biomarkers (MDA, GPX, and SOD) in the prefrontal cortex and hippocampus, induce anxiety, and impair spatial learning and memory function. Treatment with 4-MU markedly reduced anxiety levels and improved spatial learning and memory dysfunction via restoring the antioxidant defense biomarkers to normal values and reducing MDA levels. Moreover, more intact cells with normal morphologies were detected in STR-induced animals treated with 4-MU. 4-MU could attenuate the STR-induced anxiety-like behaviors and spatial learning and memory dysfunction by reducing oxidative damage and neuronal loss in the prefrontal cortex and hippocampus region. Taken together, our findings provide new insights regarding the potential therapeutic effects of 4-MU against neurobehavioral disorders induced by STR.
Collapse
Affiliation(s)
| | - Gholam Hossein Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Wang R, Yang X, Wang L, Wang R, Zhang W, Ji Y, Li Z, Li H, Cui L. β-Hydroxybutyrate alleviates brain aging through the MTA1 pathway in D-galactose injured mice. Eur J Pharmacol 2024; 983:176959. [PMID: 39216746 DOI: 10.1016/j.ejphar.2024.176959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Aging is an inevitable law of the process of life during which many physiological functions change. Brain aging is an important mechanism in the occurrence and development of degenerative diseases of the central nervous system. β-Hydroxybutyrate (BHBA) is a water-soluble, endogenous small-molecule ketone that can cross the blood-brain barrier and induce neuroprotective effects. This study aimed to investigate the effects of BHBA on D-galactose (D-gal) induced aging in mice and its underlying mechanisms using in vitro and in vivo experiments. These results indicated that D-gal-induced senescence, oxidative stress, and inflammatory responses were inhibited by BHBA, and autophagy was promoted by BHBA. Mechanistically, we explored the role of metastasis-associated antigen-1 (MTA1) in D-gal-induced damaged in HT22 cells using small interfering RNA (siRNA). The results demonstrated that the expression of MTA1 was significantly increased by BHBA, which attenuated D-gal-induced aging, oxidative stress, and inflammatory responses, and promoted autophagy through the upregulation of MTA1. In conclusion, MTA1 may be a novel target for treating aging caused by neurological damage. BHBA improves brain aging by activating the MTA1 pathway.
Collapse
Affiliation(s)
- Ruonan Wang
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, China; The First People's Hospital of Foshan, Foshan, Guangdong province, China
| | - Xiaojing Yang
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, China
| | - Li Wang
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, China
| | - Rui Wang
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, China
| | - Wanzi Zhang
- College of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, Liaoning province, China
| | - Yu Ji
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, China
| | - Zaiyu Li
- Heyou Hospital of Guangdong Province.
| | - Hua Li
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, China.
| | - Lianxu Cui
- The First People's Hospital of Foshan, Foshan, Guangdong province, China.
| |
Collapse
|
3
|
Li N, Zhang Z, Shen L, Song G, Tian J, Liu Q, Ni J. Selenium metabolism and selenoproteins function in brain and encephalopathy. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-023-2621-7. [PMID: 39546178 DOI: 10.1007/s11427-023-2621-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/09/2024] [Indexed: 11/17/2024]
Abstract
Selenium (Se) is an essential trace element of the utmost importance to human health. Its deficiency induces various disorders. Se species can be absorbed by organisms and metabolized to hydrogen selenide for the biosynthesis of selenoproteins, selenonucleic acids, or selenosugars. Se in mammals mainly acts as selenoproteins to exert their biological functions. The brain ranks highest in the specific hierarchy of organs to maintain the level of Se and the expression of selenoproteins under the circumstances of Se deficiency. Dyshomeostasis of Se and dysregulation of selenoproteins result in encephalopathy such as Alzheimer's disease, Parkinson's disease, depression, amyotrophic lateral sclerosis, and multiple sclerosis. This review provides a summary and discussion of Se metabolism, selenoprotein function, and their roles in modulating brain diseases based on the most currently published literature. It focuses on how Se is utilized and transported to the brain, how selenoproteins are biosynthesized and function physiologically in the brain, and how selenoproteins are involved in neurodegenerative diseases. At the end of this review, the perspectives and problems are outlined regarding Se and selenoproteins in the regulation of encephalopathy.
Collapse
Affiliation(s)
- Nan Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Zhonghao Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| | - Liming Shen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Guoli Song
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| | - Jing Tian
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China.
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China.
| | - Jiazuan Ni
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| |
Collapse
|
4
|
Chidambaram SB, Anand N, Varma SR, Ramamurthy S, Vichitra C, Sharma A, Mahalakshmi AM, Essa MM. Superoxide dismutase and neurological disorders. IBRO Neurosci Rep 2024; 16:373-394. [PMID: 39007083 PMCID: PMC11240301 DOI: 10.1016/j.ibneur.2023.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/21/2023] [Indexed: 07/16/2024] Open
Abstract
Superoxide dismutase (SOD) is a common antioxidant enzyme found majorly in living cells. The main physiological role of SOD is detoxification and maintain the redox balance, acts as a first line of defence against Reactive nitrogen species (RNS), Reactive oxygen species (ROS), and other such potentially hazardous molecules. SOD catalyses the conversion of superoxide anion free radicals (O 2 -.) into molecular oxygen (O 2) and hydrogen peroxide (H 2O 2) in the cells. Superoxide dismutases (SODs) are expressed in neurons and glial cells throughout the CNS both intracellularly and extracellularly. Endogenous oxidative stress (OS) linked with enlarged production of reactive oxygen metabolites (ROMs), inflammation, deregulation of redox balance, mitochondrial dysfunction and bioenergetic crisis are found to be prerequisite for neuronal loss in neurological diseases. Clinical and genetic studies indicate a direct correlation between mutations in SOD gene and neurodegenerative diseases, like Amyotrophic Lateral Sclerosis (ALS), Huntington's disease (HD), Parkinson's Disease (PD) and Alzheimer's Disease (AD). Therefore, inhibitors of OS are considered as an optimistic approach to prevent neuronal loss. SOD mimetics like Metalloporphyrin Mn (II)-cyclic polyamines, Nitroxides and Mn (III)- Salen complexes are designed and used as therapeutic extensively in the treatment of neurological disorders. SODs and SOD mimetics are promising future therapeutics in the field of various diseases with OS-mediated pathology.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Nikhilesh Anand
- Department of Pharmacology, American University of Antigua College of Medicine, University Park, Jabberwock Beach Road, Antigua, Antigua and Barbuda
| | - Sudhir Rama Varma
- Department of Clinical Sciences, College of Dentistry, Ajman University, 346 Ajman, the United Arab Emirates
- Center of Medical and Bio-allied Health Sciences Research, Ajman University, 346 Ajman, the United Arab Emirates
| | - Srinivasan Ramamurthy
- College of Pharmacy & Health Sciences, University of Science and Technology of Fujairah, 2202 Fujairah, the United Arab Emirates
| | - Chandrasekaran Vichitra
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Ambika Sharma
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
5
|
Xu Y, Xue M, Li J, Ma Y, Wang Y, Zhang H, Liang H. Fucoidan Improves D-Galactose-Induced Cognitive Dysfunction by Promoting Mitochondrial Biogenesis and Maintaining Gut Microbiome Homeostasis. Nutrients 2024; 16:1512. [PMID: 38794753 PMCID: PMC11124141 DOI: 10.3390/nu16101512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Recent studies have indicated that fucoidan has the potential to improve cognitive impairment. The objective of this study was to demonstrate the protective effect and possible mechanisms of fucoidan in D-galactose (D-gal)-induced cognitive dysfunction. Sprague Dawley rats were injected with D-galactose (200 mg/kg, sc) and administrated with fucoidan (100 mg/kg or 200 mg/kg, ig) for 8 weeks. Our results suggested that fucoidan significantly ameliorated cognitive impairment in D-gal-exposed rats and reversed histopathological changes in the hippocampus. Fucoidan reduced D-gal-induced oxidative stress, declined the inflammation level and improved mitochondrial dysfunction in hippocampal. Fucoidan promoted mitochondrial biogenesis by regulating the PGC-1α/NRF1/TFAM pathway, thereby improving D-gal-induced mitochondrial dysfunction. The regulation effect of fucoidan on PGC-1α is linked to the upstream protein of APN/AMPK/SIRT1. Additionally, the neuroprotective action of fucoidan could be related to maintaining intestinal flora homeostasis with up-regulation of Bacteroidota, Muribaculaceae and Akkermansia and down-regulation of Firmicutes. In summary, fucoidan may be a natural, promising candidate active ingredient for age-related cognitive impairment interventions.
Collapse
Affiliation(s)
- Yan Xu
- School of Public Health, Qingdao University, Qingdao 266071, China; (Y.X.); (J.L.); (Y.M.); (Y.W.); (H.Z.)
| | - Meilan Xue
- Basic Medical College, Qingdao University, Qingdao 266071, China;
| | - Jing Li
- School of Public Health, Qingdao University, Qingdao 266071, China; (Y.X.); (J.L.); (Y.M.); (Y.W.); (H.Z.)
| | - Yiqing Ma
- School of Public Health, Qingdao University, Qingdao 266071, China; (Y.X.); (J.L.); (Y.M.); (Y.W.); (H.Z.)
| | - Yutong Wang
- School of Public Health, Qingdao University, Qingdao 266071, China; (Y.X.); (J.L.); (Y.M.); (Y.W.); (H.Z.)
| | - Huaqi Zhang
- School of Public Health, Qingdao University, Qingdao 266071, China; (Y.X.); (J.L.); (Y.M.); (Y.W.); (H.Z.)
| | - Hui Liang
- School of Public Health, Qingdao University, Qingdao 266071, China; (Y.X.); (J.L.); (Y.M.); (Y.W.); (H.Z.)
| |
Collapse
|
6
|
Kanika, Singh L. Mitigating cognitive deficits with teriflunomide: unraveling PI3K-modulated behavioral outcomes in mice. Mol Biol Rep 2024; 51:572. [PMID: 38722394 DOI: 10.1007/s11033-024-09502-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/01/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Alzheimer's disease is a leading neurological disorder that gradually impairs memory and cognitive abilities, ultimately leading to the inability to perform even basic daily tasks. Teriflunomide is known to preserve neuronal activity and protect mitochondria in the brain slices exposed to oxidative stress. The current research was undertaken to investigate the teriflunomide's cognitive rescuing abilities against scopolamine-induced comorbid cognitive impairment and its influence on phosphatidylinositol-3-kinase (PI3K) inhibition-mediated behavior alteration in mice. METHODS Swiss albino mice were divided into 7 groups; vehicle control, scopolamine, donepezil + scopolamine, teriflunomide (10 mg/kg) + scopolamine; teriflunomide (20 mg/kg) + scopolamine, LY294002 and LY294002 + teriflunomide (20 mg/kg). Mice underwent a nine-day protocol, receiving scopolamine injections (2 mg/kg) for the final three days to induce cognitive impairment. Donepezil, teriflunomide, and LY294002 treatments were given continuously for 9 days. MWM, Y-maze, OFT and rota-rod tests were conducted on days 7 and 9. On the last day, blood samples were collected for serum TNF-α analysis, after which the mice were sacrificed, and brain samples were harvested for oxidative stress analysis. RESULTS Scopolamine administration for three consecutive days increased the time required to reach the platform in the MWM test, whereas, reduced the percentage of spontaneous alternations in the Y-maze, number of square crossing in OFT and retention time in the rota-rod test. In biochemical analysis, scopolamine downregulated the brain GSH level, whereas it upregulated the brain TBARS and serum TNF-α levels. Teriflunomide treatment effectively mitigated all the behavioral and biochemical alterations induced by scopolamine. Furthermore, LY294002 administration reduced the memory function and GSH level, whereas, uplifted the serum TNF-α levels. Teriflunomide abrogated the memory-impairing, GSH-lowering, and TNF-α-increasing effects of LY294002. CONCLUSION Our results delineate that the improvement in memory, locomotion, and motor coordination might be attributed to the oxidative and inflammatory stress inhibitory potential of teriflunomide. Moreover, PI3K inhibition-induced memory impairment might be attributed to reduced GSH levels and increased TNF-α levels.
Collapse
Affiliation(s)
- Kanika
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India
| | - Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India.
| |
Collapse
|
7
|
Jarosz ŁS, Socała K, Michalak K, Wiater A, Ciszewski A, Majewska M, Marek A, Grądzki Z, Wlaź P. The effect of psychoactive bacteria, Bifidobacterium longum Rosell®-175 and Lactobacillus rhamnosus JB-1, on brain proteome profiles in mice. Psychopharmacology (Berl) 2024; 241:925-945. [PMID: 38156998 PMCID: PMC11031467 DOI: 10.1007/s00213-023-06519-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
RATIONALE The gut microbiota may play an important role in the development and functioning of the mammalian central nervous system. The assumption of the experiment was to prove that the use of probiotic bacterial strains in the diet of mice modifies the expression of brain proteins involved in metabolic and immunological processes. OBJECTIVES AND RESULTS Albino Swiss mice were administered with Bifidobacterium longum Rosell®-175 or Lactobacillus rhamnosus JB-1 every 24 h for 28 days. Protein maps were prepared from hippocampal homogenates of euthanized mice. Selected proteins that were statistically significant were purified and concentrated and identified using MALDI-TOF mass spectrometry. Among the analysed samples, 13 proteins were identified. The mean volumes of calcyon, secreted frizzled-associated protein 3, and catalase in the hippocampus of mice from both experimental groups were statistically significantly higher than in the control group. In mice supplemented with Lactobacillus rhamnosus JB-1, a lower mean volume of fragrance binding protein 2, shadow of prion protein, and glycine receptor α4 subunit was observed compared to the control. CONCLUSION The psychobiotics Bifidobacterium longum Rosell®-175 and Lactobacillus rhamnosus JB-1enhances expression of proteins involved in the activation and maturation of nerve cells, as well as myelination and homeostatic regulation of neurogenesis in mice. The tested psychobiotics cause a decrease in the expression of proteins associated with CNS development and in synaptic transmission, thereby reducing the capacity for communication between nerve cells. The results of the study indicate that psychobiotic bacteria can be used in auxiliary treatment of neurological disorders.
Collapse
Affiliation(s)
- Łukasz S Jarosz
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612, Lublin, Poland.
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Katarzyna Michalak
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612, Lublin, Poland
| | - Adrian Wiater
- Department of Industrial and Environmental Microbiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Artur Ciszewski
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612, Lublin, Poland
| | - Małgorzata Majewska
- Department of Industrial and Environmental Microbiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Agnieszka Marek
- Department of Preventive Veterinary and Avian Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612, Lublin, Poland
| | - Zbigniew Grądzki
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612, Lublin, Poland
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| |
Collapse
|
8
|
Kakizawa S, Arasaki T, Yoshida A, Sato A, Takino Y, Ishigami A, Akaike T, Yanai S, Endo S. Essential role of ROS - 8-Nitro-cGMP signaling in long-term memory of motor learning and cerebellar synaptic plasticity. Redox Biol 2024; 70:103053. [PMID: 38340634 PMCID: PMC10869263 DOI: 10.1016/j.redox.2024.103053] [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: 06/05/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Although reactive oxygen species (ROS) are known to have harmful effects in organisms, recent studies have demonstrated expression of ROS synthases at various parts of the organisms and the controlled ROS generation, suggesting possible involvement of ROS signaling in physiological events of individuals. However, physiological roles of ROS in the CNS, including functional roles in higher brain functions or neuronal activity-dependent ROS production, remain to be elucidated. Here, we demonstrated involvement of ROS - 8-NO2-cGMP signaling in motor learning and synaptic plasticity in the cerebellum. In the presence of inhibitors of ROS signal or ROS synthases, cerebellar motor learning was impaired, and the stimulus inducing long-term depression (LTD), cellular basis for the motor learning, failed to induce LTD but induced long-term potentiation (LTP)-like change at cerebellar synapses. Furthermore, ROS was produced by LTD-inducing stimulus in enzyme-dependent manner, and excess administration of the antioxidant vitamin E impaired cerebellar motor learning, suggesting beneficial roles of endogenous ROS in the learning. As a downstream signal, involvement of 8-NO2-cGMP in motor learning and cerebellar LTD were also revealed. These findings indicate that ROS - 8-NO2-cGMP signal is activated by neuronal activity and is essential for cerebellum-dependent motor learning and synaptic plasticity, demonstrating involvement of the signal in physiological function of brain systems.
Collapse
Affiliation(s)
- Sho Kakizawa
- Department of Biological Chemistry, Graduate School and Faculty of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan.
| | - Tomoko Arasaki
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Ayano Yoshida
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Ayami Sato
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Yuka Takino
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan.
| |
Collapse
|
9
|
Kakizawa S, Park JJ, Tonoki A. Biology of cognitive aging across species. Geriatr Gerontol Int 2024; 24 Suppl 1:15-24. [PMID: 38126240 DOI: 10.1111/ggi.14782] [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: 08/31/2023] [Revised: 11/27/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023]
Abstract
Aging is associated with cognitive decline, which can critically affect quality of life. Examining the biology of cognitive aging across species will lead to a better understanding of the fundamental mechanisms involved in this process, and identify potential interventions that could help to improve cognitive function in aging individuals. This minireview aimed to explore the mechanisms and processes involved in cognitive aging across a range of species, from flies to rodents, and covers topics, such as the role of reactive oxygen species and autophagy/mitophagy in cognitive aging. Overall, this literature provides a comprehensive overview of the biology of cognitive aging across species, highlighting the latest research findings and identifying potential avenues for future research. Geriatr Gerontol Int 2024; 24: 15-24.
Collapse
Affiliation(s)
- Sho Kakizawa
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Joong-Jean Park
- Department of Physiology, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ayako Tonoki
- Department of Biochemistry, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
10
|
Sadeghzadeh J, Hosseini L, Mobed A, Zangbar HS, Jafarzadeh J, Pasban J, Shahabi P. The Impact of Cerebral Ischemia on Antioxidant Enzymes Activity and Neuronal Damage in the Hippocampus. Cell Mol Neurobiol 2023; 43:3915-3928. [PMID: 37740074 DOI: 10.1007/s10571-023-01413-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/09/2023] [Indexed: 09/24/2023]
Abstract
Cerebral ischemia and subsequent reperfusion, leading to reduced blood supply to specific brain areas, remain significant contributors to neurological damage, disability, and mortality. Among the vulnerable regions, the subcortical areas, including the hippocampus, are particularly susceptible to ischemia-induced injuries, with the extent of damage influenced by the different stages of ischemia. Neural tissue undergoes various changes and damage due to intricate biochemical reactions involving free radicals, oxidative stress, inflammatory responses, and glutamate toxicity. The consequences of these processes can result in irreversible harm. Notably, free radicals play a pivotal role in the neuropathological mechanisms following ischemia, contributing to oxidative stress. Therefore, the function of antioxidant enzymes after ischemia becomes crucial in preventing hippocampal damage caused by oxidative stress. This study explores hippocampal neuronal damage and enzymatic antioxidant activity during ischemia and reperfusion's early and late stages.
Collapse
Affiliation(s)
- Jafar Sadeghzadeh
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Leila Hosseini
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Ahmad Mobed
- Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Hamid Soltani Zangbar
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Jaber Jafarzadeh
- Department of Community Nutrition Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Jamshid Pasban
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Parviz Shahabi
- Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran.
| |
Collapse
|
11
|
Liu H, Inoue R, Koyanagi M, Hayashi SM, Nagaoka K. Potential Effects of Alpha-Glycosyl Isoquercitrin on Memory by Altering the Gut Microbiota-Blood-Brain Axis in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15991-16002. [PMID: 37861708 DOI: 10.1021/acs.jafc.3c00897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Alpha-glycosyl isoquercitrin (AGIQ), composed of isoquercitrin and glycosylated quercetin, has multiple biological effects. Here, we further examined the influence of AGIQ on brain function and provided its potential mechanism. Male C57BL/6 mice were treated with 0, 0.005, and 0.05% AGIQ in drinking water for 4 weeks prior to behavioral testing. Behavior tests showed that 0.05% AGIQ treatment significantly improved learning and memory function without affecting emotion. In the hippocampus, the gene expression of antioxidative defense enzymes was upregulated after 0.05% AGIQ treatment. In contrast, AGIQ caused significant alterations in the microbial abundance of genera Akkermansia, Bifidobacterium, and Alistipes associated with memory function. Metabolomics analysis identified that taurine concentration was significantly increased in serum and hippocampus from AGIQ-treated mice. The correlation analysis suggested that elevated serum taurine levels were closely related to the abundance of Akkermansia, indicating the underlying crosstalk of gut microbiota and serum metabolites. In vitro fecal culture further demonstrated that AGIQ could increase the level of Akkermansia. Taurine could exert antioxidant activity in SH-SY5Y neuroblastoma cell lines in vitro. Finally, vancomycin-induced alterations of gut microbiota attenuated the taurine increases in the serum and the antioxidant gene level in the hippocampus by AGIQ. Taken together, it is likely that AGIQ could increase genus Akkermansia abundance and ultimately increase taurine levels in serum and hippocampus to improve learning and memory function, relying on the gut microbiota-blood-brain axis. Our results supply a new view for understanding effects of AGIQ on brain function.
Collapse
Affiliation(s)
- Hong Liu
- Laboratory of Veterinary Physiology, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Setsunan University, Osaka 573-0101, Japan
| | | | - Shim-Mo Hayashi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
- Division of Food Additives, National Institute of Health Sciences, Kawasaki 210-0821, Kanagawa, Japan
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| |
Collapse
|
12
|
Ly M, Yu GZ, Mian A, Cramer A, Meysami S, Merrill DA, Samara A, Eisenstein SA, Hershey T, Babulal GM, Lenze EJ, Morris JC, Benzinger TLS, Raji CA. Neuroinflammation: A Modifiable Pathway Linking Obesity, Alzheimer's disease, and Depression. Am J Geriatr Psychiatry 2023; 31:853-866. [PMID: 37365110 PMCID: PMC10528955 DOI: 10.1016/j.jagp.2023.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023]
Abstract
Obesity, depression and Alzheimer's disease (AD) are three major interrelated modern health conditions with complex relationships. Early-life depression may serve as a risk factor for AD, while late-life depression may be a prodrome of AD. Depression affects approximately 23% of obese individuals, and depression itself raises the risk of obesity by 37%. Mid-life obesity independently increases AD risk, while late-life obesity, particularly metabolically healthy obesity, may offer protection against AD pathology. Chronic inflammation serves as a key mechanism linking obesity, AD, and depression, encompassing systemic inflammation from metabolic disturbances, immune dysregulation through the gut microbiome, and direct interactions with amyloid pathology and neuroinflammation. In this review, we explore the biological mechanisms of neuroinflammation in relation to obesity, AD, and depression. We assess the efficacy of therapeutic interventions targeting neuroinflammation and discuss current and future radiological imaging initiatives for studying neuroinflammation. By comprehending the intricate interplay among depression, obesity, and AD, especially the role of neuroinflammation, we can advance our understanding and develop innovative strategies for prevention and treatment.
Collapse
Affiliation(s)
- Maria Ly
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO
| | - Gary Z Yu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO
| | - Ali Mian
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO
| | | | - Somayeh Meysami
- Pacific Brain Health Center, Pacific Neuroscience Institute Foundation, Santa Monica, CA; Department of Translational Neurosciences, Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA
| | - David A Merrill
- Pacific Brain Health Center, Pacific Neuroscience Institute Foundation, Santa Monica, CA; Department of Translational Neurosciences, Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA
| | - Amjad Samara
- Department of Neurology, Washington University in St. Louis, St. Louis, MO
| | - Sarah A Eisenstein
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO; Department of Psychiatry, Washington University in St. Louis, St. Louis, MO
| | - Tamara Hershey
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO; Department of Psychological & Brain Sciences, Washington University School of Medicine, St. Louis, MO
| | - Ganesh M Babulal
- Department of Neurology, Washington University in St. Louis, St. Louis, MO; Institute of Public Health, Washington University in St. Louis, St. Louis, MO; Department of Psychology, Faculty of Humanities, University of Johannesburg, Johannesburg, South Africa; Department of Clinical Research and Leadership, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Eric J Lenze
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO
| | - John C Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, MO
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO; Department of Neurological Surgery, Washington University in St. Louis, St. Louis, MO
| | - Cyrus A Raji
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO; Department of Neurology, Washington University in St. Louis, St. Louis, MO.
| |
Collapse
|
13
|
Veselov IM, Vinogradova DV, Maltsev AV, Shevtsov PN, Spirkova EA, Bachurin SO, Shevtsova EF. Mitochondria and Oxidative Stress as a Link between Alzheimer's Disease and Diabetes Mellitus. Int J Mol Sci 2023; 24:14450. [PMID: 37833898 PMCID: PMC10572926 DOI: 10.3390/ijms241914450] [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: 08/14/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
This review is devoted to the problems of the common features linking metabolic disorders and type 2 diabetes with the development of Alzheimer's disease. The pathogenesis of Alzheimer's disease closely intersects with the mechanisms of type 2 diabetes development, and an important risk factor for both pathologies is aging. Common pathological mechanisms include both factors in the development of oxidative stress, neuroinflammation, insulin resistance, and amyloidosis, as well as impaired mitochondrial dysfunctions and increasing cell death. The currently available drugs for the treatment of type 2 diabetes and Alzheimer's disease have limited therapeutic efficacy. It is important to note that drugs used to treat Alzheimer's disease, in particular acetylcholinesterase inhibitors, show a positive therapeutic potential in the treatment of type 2 diabetes, while drugs used in the treatment of type 2 diabetes can also prevent a number of pathologies characteristic for Alzheimer's disease. A promising direction in the search for a strategy for the treatment of type 2 diabetes and Alzheimer's disease may be the creation of complex multi-target drugs that have neuroprotective potential and affect specific common targets for type 2 diabetes and Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Elena F. Shevtsova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences (IPAC RAS), Chernogolovka 142432, Russia; (I.M.V.); (A.V.M.); (P.N.S.); (E.A.S.); (S.O.B.)
| |
Collapse
|
14
|
Levings DC, Pathak SS, Yang YM, Slattery M. Limited expression of Nrf2 in neurons across the central nervous system. Redox Biol 2023; 65:102830. [PMID: 37544245 PMCID: PMC10428127 DOI: 10.1016/j.redox.2023.102830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/08/2023] Open
Abstract
Nrf2, encoded by the gene Nfe2l2, is a broadly expressed transcription factor that regulates gene expression in response to reactive oxygen species (ROS) and oxidative stress. It is commonly referred to as a ubiquitous pathway, but this generalization overlooks work indicating that Nrf2 is essentially unexpressed in some neuronal populations. To explore whether this pattern extends throughout the central nervous system (CNS), we quantified Nfe2l2 expression and chromatin accessibility at the Nfe2l2 locus across multiple single cell datasets. In both the mouse and human CNS, Nfe2l2 was repressed in almost all mature neurons, but highly expressed in non-neuronal support cells, and this pattern was robust across multiple human CNS diseases. A subset of key Nrf2 target genes, like Slc7a11, also remained low in neurons. Thus, these data suggest that while most cells express Nfe2l2, with activity determined by ROS levels, neurons actively avoid Nrf2 activity by keeping Nfe2l2 expression low.
Collapse
Affiliation(s)
- Daniel C Levings
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA
| | - Salil Saurav Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA; Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Matthew Slattery
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA.
| |
Collapse
|
15
|
Rahman MA, Shuvo AA, Apu MMH, Bhakta MR, Islam F, Rahman MA, Islam MR, Reza HM. Combination of epigallocatechin 3 gallate and curcumin improves D-galactose and normal-aging associated memory impairment in mice. Sci Rep 2023; 13:12681. [PMID: 37542120 PMCID: PMC10403524 DOI: 10.1038/s41598-023-39919-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/02/2023] [Indexed: 08/06/2023] Open
Abstract
Previously, we observed curcumin improves aging-associated memory impairment in D-galactose (D-gal) and normal-aged (NA) mice. Evidence showed that multiple agents can be used in managing aging-induced memory dysfunction, drawn by the contribution of several pathways. Curcumin and Epigallocatechin 3 gallate (EGCG) combination substantially reduced the oxidative stress that commonly mediates aging. This study examined the combined effect of EGCG and curcumin on memory improvement in two recognized models, D-gal and normal-aged (NA) mice. The co-administration of EGCG and curcumin significantly (p < 0.05) increased retention time detected by passive avoidance (PA) and freezing response determined in contextual fear conditioning (CFC) compared to the discrete administration of EGCG or curcumin. Biochemical studies revealed that the combination of EGCG and curcumin remarkably ameliorated the levels (p < 0.05) of glutathione, superoxide dismutase, catalase, advanced oxidation protein products, nitric oxide, and lipid peroxidation compared to the monotherapy of EGCG or curcumin in mice hippocampi. The behavioral and biochemical studies revealed that the combination of EGCG and curcumin showed better improvement in rescuing aging-associated memory disorders in mice. EGCG and curcumin combination could serve as a better choice in managing aging-related memory disorders.
Collapse
Affiliation(s)
- Md Ashrafur Rahman
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh.
- Department of Pharmaceutical Sciences, Wilkes University, Wilkes Barre, PA, 18766, USA.
| | - Arif Anzum Shuvo
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| | - Md Mehedi Hasan Apu
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| | - Monisha Rani Bhakta
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| | - Farzana Islam
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| | - Md Atiqur Rahman
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| | - Md Rabiul Islam
- Department of Pharmacy, University of Asia Pacific, 74/A Green Road, Farmgate, Dhaka, 1205, Bangladesh.
- School of Pharmacy, BRAC University, 66 Mohakhali, Dhaka, 1212, Bangladesh.
| | - Hasan Mahmud Reza
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh.
| |
Collapse
|
16
|
Hajipour S, Farbood Y, Dianat M, Nesari A, Sarkaki A. Effect of Berberine against Cognitive Deficits in Rat Model of Thioacetamide-Induced Liver Cirrhosis and Hepatic Encephalopathy (Behavioral, Biochemical, Molecular and Histological Evaluations). Brain Sci 2023; 13:944. [PMID: 37371422 DOI: 10.3390/brainsci13060944] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/17/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Liver cirrhosis (LC) is one of the chronic liver diseases with high disability and mortality accompanying hepatic encephalopathy (HE) followed by cognitive dysfunctions. In this work, the effect of berberine (Ber) on spatial cognition was studied in a rat model of LC induced by thioacetamide (TAA). MATERIALS AND METHODS Male Wistar rats (200-250 g) were divided into six groups: (1) control; (2) TAA, 200 mg/kg/day, i.p.; (3-5) TAA + Ber; received Ber (10, 30, and 60 mg/kg, i.p., daily after last TAA injection); (6) Dizocilpine (MK-801) + TAA, received MK-801 (2 mg/kg/day, i.p.) 30 m before TAA injection. The spatial memory, BBB permeability, brain edema, liver enzymes, urea, serum and brain total bilirubin, oxidative stress and cytokine markers in the hippocampus were measured. Furthermore, a histological examination of the hippocampus was carried out. RESULTS The BBB permeability, brain edema, liver enzymes, urea, total bilirubin levels in serum and hippocampal MDA and TNF-α increased significantly after TAA injection (p < 0.001); the spatial memory was impaired (p < 0.001), and hippocampal IL-10 decreased (p < 0.001). Ber reversed all the above parameters significantly (p < 0.05, p < 0.01 and p < 0.001). MK-801 prevented the development of LC via TAA (p < 0.001). CONCLUSION Results showed that Ber improves spatial learning and memory in TAA-induced LC by improving the BBB function, oxidative stress and neuroinflammation. Ber might be a promising therapeutic agent for cognitive improvement in LC.
Collapse
Affiliation(s)
- Somayeh Hajipour
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
| | - Yaghoob Farbood
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
- Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
| | - Mahin Dianat
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
- Department of Physiology, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
| | - Ali Nesari
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
| | - Alireza Sarkaki
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
- Medicinal Plants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz P.O. Box 61355-15795, Iran
- National Institute for Medical Research Development "NIMAD", Tehran 1419693111, Iran
| |
Collapse
|
17
|
Levings DC, Pathak SS, Yang YM, Slattery M. Limited Expression of Nrf2 in Neurons Across the Central Nervous System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.09.540014. [PMID: 37214946 PMCID: PMC10197674 DOI: 10.1101/2023.05.09.540014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Nrf2 is a broadly expressed transcription factor that regulates gene expression in response to reactive oxygen species (ROS) and oxidative stress. It is commonly referred to as a ubiquitous pathway, but this generalization overlooks work indicating that Nrf2 is essentially unexpressed in some neuronal populations. To explore whether this pattern extends throughout the central nervous system (CNS), we quantified Nrf2 expression and chromatin accessibility at the Nrf2 locus across multiple single cell datasets. In both the mouse and human CNS, Nrf2 was repressed in almost all mature neurons, but highly expressed in non-neuronal support cells, and this pattern was robust across multiple human CNS diseases. A subset of key Nrf2 target genes, like Slc7a11 , also remained low in neurons. Thus, these data suggest that while most cells express Nrf2, with activity determined by ROS levels, neurons actively avoid Nrf2 activity by keeping Nrf2 expression low.
Collapse
Affiliation(s)
- Daniel C. Levings
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Salil Saurav Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Matthew Slattery
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| |
Collapse
|
18
|
Carvalho C, Moreira PI. Metabolic defects shared by Alzheimer's disease and diabetes: A focus on mitochondria. Curr Opin Neurobiol 2023; 79:102694. [PMID: 36842275 DOI: 10.1016/j.conb.2023.102694] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/26/2023]
Abstract
Type 2 diabetes (T2D) and Alzheimer's disease (AD) are two global epidemics that share several metabolic defects, such as insulin resistance, impaired glucose metabolism, and mitochondrial defects. Importantly, strong evidence demonstrates that T2D significantly increases the risk of cognitive decline and dementia, particularly AD. Here, we provide an overview of the metabolic defects that characterize and link both pathologies putting the focus on mitochondria. The biomarker potential of mitochondrial components and the therapeutic potential of some drugs that target and modulate mitochondria are also briefly discussed.
Collapse
Affiliation(s)
- Cristina Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Center for Innovation in Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Center for Innovation in Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
19
|
Thongrong S, Surapinit S, Promsrisuk T, Jittiwat J, Kongsui R. Pinostrobin alleviates chronic restraint stress‑induced cognitive impairment by modulating oxidative stress and the function of astrocytes in the hippocampus of rats. Biomed Rep 2023; 18:20. [PMID: 36798091 PMCID: PMC9922797 DOI: 10.3892/br.2023.1602] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Chronic stress has been recognized to induce the alterations of neuronal and glial cells in the hippocampus, and is thus implicated in cognitive dysfunction. There is increasing evidence to indicate that natural compounds capable of exerting neuroprotective and antioxidant activities, may function as potential therapeutic agents for cognitive impairment. The present study examined the neuroprotective effects of pinostrobin from Boesenbergia rotunda (L.) against chronic restraint stress (CRS)-induced cognitive impairment associated with the alterations of oxidative stress, neuronal density and glial fibrillary acidic protein (GFAP) of astrocytes in the hippocampus. For this purpose, male Wistar rats were administered once daily with pinostrobin (20 and 40 mg/kg, per os) prior to exposure to CRS (6 h/day) for 21 days. The cognitive behaviors, the concentration of malondialdehyde, and the activities of superoxide dismutase and catalase were determined. Histologically, the alterations in astrocytic GFAP and excitatory amino acid transporter 2 (EAAT2) in the hippocampus were examined. The results revealed that pinostrobin potentially attenuated cognitive impairment in the Y-maze and in novel object recognition tests, with a reduction in oxidative stress. Furthermore, pinostrobin effectively increased neuronal density, as well as the immunoreactivities of GFAP and EAAT2 in the hippocampus. Taken together, these findings indicate that treatment with pinostrobin alleviates chronic stress-induced cognitive impairment by exerting antioxidant effects, reducing neuronal cell damage, and improving the function of astrocytic GFAP and EAAT2. Thus, pinostrobin may have potential for use as a neuroprotective agent to protect against chronic stress-induced brain dysfunction and cognitive deficits.
Collapse
Affiliation(s)
- Sitthisak Thongrong
- Division of Anatomy, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand,Unit of Excellence in Translational Neurosciences Initiative, University of Phayao, Phayao 56000, Thailand
| | - Serm Surapinit
- Unit of Excellence in Translational Neurosciences Initiative, University of Phayao, Phayao 56000, Thailand,Department of Medical Technology, School of Allied Health Sciences, University of Phayao, Phayao 56000, Thailand
| | - Tichanon Promsrisuk
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Jinatta Jittiwat
- Faculty of Medicine, Mahasarakham University, Mahasarakham 44000, Thailand
| | - Ratchaniporn Kongsui
- Unit of Excellence in Translational Neurosciences Initiative, University of Phayao, Phayao 56000, Thailand,Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand,Correspondence to: Dr Ratchaniporn Kongsui, Division of Physiology, School of Medical Sciences, University of Phayao, 19 Moo 2 Phahonyothin Road, Maeka, Muang Phayao, Phayao 56000, Thailand
| |
Collapse
|
20
|
Lin S, Chen C, Ouyang P, Cai Z, Liu X, Abdurahman A, Peng J, Li Y, Zhang Z, Song GL. SELENOM Knockout Induces Synaptic Deficits and Cognitive Dysfunction by Influencing Brain Glucose Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1607-1619. [PMID: 36635091 DOI: 10.1021/acs.jafc.2c07491] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Selenium, a trace element associated with memory impairment and glucose metabolism, mainly exerts its function through selenoproteins. SELENOM is a selenoprotein located in the endoplasmic reticulum (ER) lumen. Our study demonstrates for the first time that SELENOM knockout decreases synaptic plasticity and causes memory impairment in 10-month-old mice. In addition, SELENOM knockout causes hyperglycaemia and disturbs glucose metabolism, which is essential for synapse formation and transmission in the brain. Further research reveals that SELENOM knockout leads to inhibition of the brain insulin signaling pathway [phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR/p70 S6 kinase pathway], which may impair synaptic plasticity in mice. High-fat diet (HFD) feeding suppresses the brain insulin signaling pathway in SELENOM knockout mice and leads to earlier onset of cognitive impairment at 5 months of age. In general, our study demonstrates that SELENOM knockout induces synaptic deficits via the brain insulin signaling pathway, thus leading to cognitive dysfunction in mice. These data strongly suggest that SELENOM plays a vital role in brain glucose metabolism and contributes substantially to synaptic plasticity.
Collapse
Affiliation(s)
- Shujing Lin
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong518000, People's Republic of China
| | - Chen Chen
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Pei Ouyang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Zhiyu Cai
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Xibei Liu
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Anwar Abdurahman
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Jiaying Peng
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Yu Li
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Zhonghao Zhang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Guo-Li Song
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
- Shenzhen Bay Laboratory, Shenzhen, Guangdong518000, People's Republic of China
| |
Collapse
|
21
|
Saito ER, Warren CE, Hanegan CM, Larsen JG, du Randt JD, Cannon M, Saito JY, Campbell RJ, Kemberling CM, Miller GS, Edwards JG, Bikman BT. A Novel Ketone-Supplemented Diet Improves Recognition Memory and Hippocampal Mitochondrial Efficiency in Healthy Adult Mice. Metabolites 2022; 12:1019. [PMID: 36355101 PMCID: PMC9693360 DOI: 10.3390/metabo12111019] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 09/13/2023] Open
Abstract
Mitochondrial dysfunction and cognitive impairment are common symptoms in many neurologic and psychiatric disorders, as well as nonpathological aging. Ketones have been suggested as therapeutic for their efficacy in epilepsy and other brain pathologies such as Alzheimer's disease and major depressive disorder. However, their effects on cognitive function in healthy individuals is less established. Here, we explored the mitochondrial and performative outcomes of a novel eight-week ketone-supplemented ketogenic (KETO) diet in healthy adult male and female mice. In a novel object recognition test, KETO mice spent more time with the novel, compared to familiar, object, indicating an improvement in recognition memory. High-resolution respirometry on permeabilized hippocampal tissue returned significant reductions in mitochondrial O2 consumption. No changes in ATP production were observed, yielding a significantly higher ATP:O2 ratio, a measure of mitochondrial efficiency. Together, these findings demonstrate the KETO diet improves hippocampal mitochondrial efficiency. They add to a growing body of evidence that suggests ketones and ketogenic diets are neuroprotective and metabolically and cognitively relevant, even in healthy adults. They also suggest that ketogenic lifestyle changes may be effective strategies for protecting against cognitive decline associated with aging and disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Benjamin T. Bikman
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
22
|
Saez-Calveras N, Stuve O. The role of the complement system in Multiple Sclerosis: A review. Front Immunol 2022; 13:970486. [PMID: 36032156 PMCID: PMC9399629 DOI: 10.3389/fimmu.2022.970486] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
The complement system has been involved in the pathogenesis of multiple neuroinflammatory and neurodegenerative conditions. In this review, we evaluated the possible role of complement activation in multiple sclerosis (MS) with a focus in progressive MS, where the disease pathogenesis remains to be fully elucidated and treatment options are limited. The evidence for the involvement of the complement system in the white matter plaques and gray matter lesions of MS stems from immunohistochemical analysis of post-mortem MS brains, in vivo serum and cerebrospinal fluid biomarker studies, and animal models of Experimental Autoimmune Encephalomyelitis (EAE). Complement knock-out studies in these animal models have revealed that this system may have a “double-edge sword” effect in MS. On the one hand, complement proteins may aid in promoting the clearance of myelin degradation products and other debris through myeloid cell-mediated phagocytosis. On the other, its aberrant activation may lead to demyelination at the rim of progressive MS white matter lesions as well as synapse loss in the gray matter. The complement system may also interact with known risk factors of MS, including as Epstein Barr Virus (EBV) infection, and perpetuate the activation of CNS self-reactive B cell populations. With the mounting evidence for the involvement of complement in MS, the development of complement modulating therapies for this condition is appealing. Herein, we also reviewed the pharmacological complement inhibitors that have been tested in MS animal models as well as in clinical trials for other neurologic diseases. The potential use of these agents, such as the C5-binding antibody eculizumab in MS will require a detailed understanding of the role of the different complement effectors in this disease and the development of better CNS delivery strategies for these compounds.
Collapse
Affiliation(s)
- Nil Saez-Calveras
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, Veterans Affairs (VA) North Texas Health Care System, Dallas, TX, United States
- *Correspondence: Olaf Stuve,
| |
Collapse
|
23
|
Prophylactic Zinc Administration Combined with Swimming Exercise Prevents Cognitive-Emotional Disturbances and Tissue Injury following a Transient Hypoxic-Ischemic Insult in the Rat. Behav Neurol 2022; 2022:5388944. [PMID: 35637877 PMCID: PMC9146809 DOI: 10.1155/2022/5388944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 11/17/2022] Open
Abstract
Exercise performance and zinc administration individually yield a protective effect on various neurodegenerative models, including ischemic brain injury. Therefore, this work was aimed at evaluating the combined effect of subacute prophylactic zinc administration and swimming exercise in a transient cerebral ischemia model. The prophylactic zinc administration (2.5 mg/kg of body weight) was provided every 24 h for four days before a 30 min common carotid artery occlusion (CCAO), and 24 h after reperfusion, the rats were subjected to swimming exercise in the Morris Water Maze (MWM). Learning was evaluated daily for five days, and memory on day 12 postreperfusion; anxiety or depression-like behavior was measured by the elevated plus maze and the motor activity by open-field test. Nitrites, lipid peroxidation, and the activity of superoxide dismutase (SOD) and catalase (CAT) were assessed in the temporoparietal cortex and hippocampus. The three nitric oxide (NO) synthase isoforms, chemokines, and their receptor levels were measured by ELISA. Nissl staining evaluated hippocampus cytoarchitecture and Iba-1 immunohistochemistry activated the microglia. Swimming exercise alone could not prevent ischemic damage but, combined with prophylactic zinc administration, reversed the cognitive deficit, decreased NOS and chemokine levels, prevented tissue damage, and increased Iba-1 (+) cell number. These results suggest that the subacute prophylactic zinc administration combined with swimming exercise, but not the individual treatment, prevents the ischemic damage on day 12 postreperfusion in the transient ischemia model.
Collapse
|
24
|
Kim MJ, Kang JY, Kim JM, Moon JH, Lee HL, Jeong HR, Go MJ, Lee U, Heo HJ. Effect of Ethyl Acetate Fraction from Eucommia ulmoides Leaves on PM 2.5-Induced Inflammation and Cognitive Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7157444. [PMID: 35607702 PMCID: PMC9124148 DOI: 10.1155/2022/7157444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/31/2022] [Accepted: 04/19/2022] [Indexed: 11/18/2022]
Abstract
This study aimed to evaluate the protective effect of the ethyl acetate from Eucommia ulmoides leaves (EFEL) on PM2.5-induced cognitive impairment in BALB/c mice. EFEL improved PM2.5-induced cognitive decline by improving spontaneous alternative behavioral and long-term memory ability. EFEL increased ferric reducing activity power (FRAP) in serum. In addition, EFEL increased superoxide dismutase (SOD) and reduced glutathione (GSH) contents and inhibited the production of malondialdehyde (MDA) in lung and brain tissues. EFEL also restored the mitochondrial function by regulating reactive oxygen species (ROS) production, mitochondrial membrane potential (MMP) level, and ATP level in lung and brain tissues. EFEL ameliorated the cholinergic system by regulating the acetylcholine (ACh) content and acetylcholinesterase (AChE) activity in the brain tissue and the expression of AChE and choline acetyltransferase (ChAT) in the whole brain and hippocampal tissues. EFEL reduced PM2.5-induced excessive expression of inflammatory protein related to the lung, whole brain, olfactory bulb, and hippocampus. Physiological compounds of EFEL were identified as 5-O-caffeolyquinic acid, rutin, quercetin, and quercetin glycosides. As a result, EFEL has anti-inflammation and anti-amnesic effect on PM2.5-induced cognitive impairment by regulating the inflammation and inhibiting the lung and brain tissue dysfunction, and its effect is considered to be due to the physiological compounds of EFEL.
Collapse
Affiliation(s)
- Min Ji Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jin Yong Kang
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
- World Institute of Kimchi an Annex of Korea Food Research Institute, Gwangju, Republic of Korea
| | - Jong Min Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jong Hyun Moon
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hyo Lim Lee
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hye Rin Jeong
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Min Ji Go
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Uk Lee
- Division of Special Forest Products, National Institute of Forest Science, Suwon 16631, Republic of Korea
| | - Ho Jin Heo
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
25
|
Ortiz GG, Huerta M, González-Usigli HA, Torres-Sánchez ED, Delgado-Lara DLC, Pacheco-Moisés FP, Mireles-Ramírez MA, Torres-Mendoza BMG, Moreno-Cih RI, Velázquez-Brizuela IE. Cognitive disorder and dementia in type 2 diabetes mellitus. World J Diabetes 2022; 13:319-337. [PMID: 35582669 PMCID: PMC9052006 DOI: 10.4239/wjd.v13.i4.319] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/14/2021] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
Insulin, a key pleiotropic hormone, regulates metabolism through several signaling pathways in target tissues including skeletal muscle, liver, and brain. In the brain, insulin modulates learning and memory, and impaired insulin signaling is associated with metabolic dysregulation and neurodegenerative diseases. At the receptor level, in aging and Alzheimer’s disease (AD) models, the amount of insulin receptors and their functions are decreased. Clinical and animal model studies suggest that memory improvements are due to changes in insulin levels. Furthermore, diabetes mellitus (DM) and insulin resistance are associated with age-related cognitive decline, increased levels of β-amyloid peptide, phosphorylation of tau protein; oxidative stress, pro-inflammatory cytokine production, and dyslipidemia. Recent evidence shows that deleting brain insulin receptors leads to mild obesity and insulin resistance without influencing brain size and apoptosis development. Conversely, deleting insulin-like growth factor 1 receptor (IGF-1R) affects brain size and development, and contributes to behavior changes. Insulin is synthesized locally in the brain and is released from the neurons. Here, we reviewed proposed pathophysiological hypotheses to explain increased risk of dementia in the presence of DM. Regardless of the exact sequence of events leading to neurodegeneration, there is strong evidence that mitochondrial dysfunction plays a key role in AD and DM. A triple transgenic mouse model of AD showed mitochondrial dysfunction, oxidative stress, and loss of synaptic integrity. These alterations are comparable to those induced in wild-type mice treated with sucrose, which is consistent with the proposal that mitochondrial alterations are associated with DM and contribute to AD development. Alterations in insulin/IGF-1 signaling in DM could lead to mitochondrial dysfunction and low antioxidant capacity of the cell. Thus, insulin/IGF-1 signaling is important for increased neural processing and systemic metabolism, and could be a specific target for therapeutic strategies to decrease alterations associated with age-related cognitive decline.
Collapse
Affiliation(s)
- Genaro G Ortiz
- Department of Philosophical and Methodological Disciplines, University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Department of Neurology, Movement Disorders Clinic, Sub-Specialty Medical Unit, National Western Medical Center, Mexican Social Security Institute (IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Miguel Huerta
- University Biomedical Research Center, University of Colima, Colima 28040, Mexico
| | - Héctor A González-Usigli
- Department of Neurology, Movement Disorders Clinic, Sub-Specialty Medical Unit, National Western Medical Center, Mexican Social Security Institute (IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Erandis D Torres-Sánchez
- Department of Medical and Life Sciences, University Center of ‘La Ciénega’, University of Guadalajara, Ocotlán 47810, Jalisco, Mexico
| | - Daniela LC Delgado-Lara
- Department of Philosophical and Methodological Disciplines, University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Fermín P Pacheco-Moisés
- Department of Chemistry, University Center of Exact Sciences and Engineering, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Mario A Mireles-Ramírez
- Department of Neurology, Movement Disorders Clinic, Sub-Specialty Medical Unit, National Western Medical Center, Mexican Social Security Institute (IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Blanca MG Torres-Mendoza
- Department of Philosophical and Methodological Disciplines, University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Division of Neurosciences, Western Biomedical Research Center, Mexican Social Security Institute (IMSS), Guadalajara 44340, Jalisco, Mexico
| | - Roxana I Moreno-Cih
- Gerontology Postgraduate Program, Public Health Department, University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Irma E Velázquez-Brizuela
- Department of Philosophical and Methodological Disciplines, University Health Sciences Center, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
26
|
Parvardeh S, Sheikholeslami MA, Ghafghazi S, Pouriran R, Mortazavi SE. Minocycline Improves Memory by Enhancing Hippocampal Synaptic Plasticity and Restoring Antioxidant Enzyme Activity in a Rat Model of Cerebral Ischemia-Reperfusion. Basic Clin Neurosci 2022; 13:225-235. [PMID: 36425949 PMCID: PMC9682322 DOI: 10.32598/bcn.12.6.2062.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/23/2021] [Accepted: 12/26/2021] [Indexed: 06/16/2023] Open
Abstract
INTRODUCTION Oxidative stress plays a crucial role in the impairment of synaptic plasticity following cerebral ischemia, ultimately resulting in memory dysfunction. Hence, the applying antioxidant agents could be beneficial in managing memory deficits after brain ischemia. Minocycline is a tetracycline antibiotic with antioxidant effect. The main objective of this work was to assess the minocycline effect on the impairment of synaptic plasticity and memory after cerebral ischemia-reperfusion in rats. METHODS Transient occlusion of common carotid arteries was used to induce ischemiareperfusion injury in rats. Single or multiple (once daily for 7 days) dose(s) of minocycline were administered before (pretreatment) or after (treatment) brain ischemia. Seven days after ischemia-reperfusion, passive avoidance performance, long-term hippocampal potentiation, and the activity of antioxidant enzymes were assessed. RESULTS The passive avoidance test showed that minocycline (20 and 40 mg/kg) significantly increased step-through latency while reducing the duration of staying in a dark chamber in the treatment (but not pretreatment) group. In electrophysiological experiments, the rats treated (but not pretreated) with minocycline (40 mg/kg) showed a significant increase in the amplitude of the field excitatory postsynaptic potentials in the dentate gyrus area of the hippocampus. The treatment (but not pretreatment) with minocycline (20 and 40 mg/kg) resulted in a significant increase in the activity of catalase, glutathione peroxidase, and superoxide dismutase in the hippocampus. CONCLUSION It was determined that minocycline attenuates memory dysfunction after cerebral ischemia-reperfusion in rats by improving hippocampal synaptic plasticity and restoring antioxidant enzyme activity. HIGHLIGHTS Minocycline enhances passive avoidance memory after cerebral ischemia-reperfusion.Minocycline increases enzymatic antioxidant capacity in hippocampal formation.Minocycline improves synaptic plasticity in perforant path-granule cell synapse. PLAIN LANGUAGE SUMMARY Stroke is a common neurological disease with a relatively high mortality rate and disabilities worldwide. More than half of the patients who have had an episode of stroke suffer from the impairment of sensorimotor function and language problems as well as learning and memory disorders. Oxidative stress plays an important role in memory impairment following brain ischemia. Hence, the application of antioxidant agents could be beneficial in managing memory deficits after stroke. Minocycline is a tetracycline antibiotic that is used for the treatment of infectious diseases; it can also function as a potent antioxidant medication. Hence, we hypothesized that minocycline could attenuate memory impairment after brain ischemia. We examined this hypothesis in a rat model of brain ischemia. In this model, the main arteries that supply the brain with oxygenated blood were occluded to induce brain ischemia in the rats. Then, minocycline was administered to the rats, which were subjected to brain ischemia. Seven days later, memory function in the rats was evaluated. The results showed that minocycline could enhance the activity of antioxidant enzymes in the brain, which physiologically fight off oxidative stress. This property of minocycline protects brain cells against ischemic injury and thereby increases the transmission of neuronal signals from one cell to another cell in the memory centers in the brain. These effects ultimately increase the memory function of rats, which was evident in the behavioral memory test. Overall, the study results suggest that minocycline can be considered a memory enhancer drug in patients who suffer from learning and memory disorders following a stroke.
Collapse
Affiliation(s)
- Siavash Parvardeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shiva Ghafghazi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramin Pouriran
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Erfan Mortazavi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Som S, Antony J, Dhanabal SP, Ponnusankar S. Neuroprotective role of Diosgenin, a NGF stimulator, against Aβ (1-42) induced neurotoxicity in animal model of Alzheimer's disease. Metab Brain Dis 2022; 37:359-372. [PMID: 35023028 DOI: 10.1007/s11011-021-00880-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 12/05/2021] [Indexed: 01/24/2023]
Abstract
Diosgenin is a neurosteroid derived from the plants and has been previously reported for its numerous health beneficial properties, such as anti-arrhythmic, hypolipidemic, and antiproliferative effects. Although several studies conducted earlier suggested cognition enhancement actions of diosgenin against neurodegenerative disorders, but the molecular mechanisms underlying are not clearly understood. In the present study, we investigated the neuroprotective effect of diosgenin in the Wistar rats that received an intracerebroventricular injection of Amyloid-β (1-42) peptides, representing a rodent model of Alzheimer's disease (AD). Animals were treated with 100 and 200 mg/kg/p.o of diosgenin for 28 days, followed by Amyloid-β (1-42) peptides infusion. Animals were assessed for the spatial learning and memory by using radial arm maze and passive avoidance task. Subsequently, animals were euthanized and brains were collected for biochemical estimations and histopathological studies. Our results revealed that, diosgenin administration dose dependently improved the spatial learning and memory and protected the animals from Amyloid-β (1-42) peptides induced disrupted cognitive functions. Further, biochemical analysis showed that diosgenin successfully attenuated Amyloid-β (1-42) mediated plaque load, oxidative stress, neuroinflammation and elevated acetylcholinesterase activity. In addition, histopathological evaluation also supported neuroprotective effects of diosgenin in hippocampus of rat brain when assessed using hematoxylin-eosin and Cresyl Violet staining. Thus, the aforementioned effects suggested protective action of diosgenin against Aβ (1-42) induced neuronal damage and thereby can serve as a potential therapeutic candidate for AD.
Collapse
Affiliation(s)
- Swati Som
- Department of Pharmacy Practice, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty-643001, Tamilnadu, India
| | - Justin Antony
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty-643001, Tamilnadu, India
| | - SPalanisamy Dhanabal
- Department of Pharmacognosy and Phytochemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty-643001, Tamilnadu, India
| | - Sivasankaran Ponnusankar
- Department of Pharmacy Practice, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty-643001, Tamilnadu, India.
| |
Collapse
|
28
|
Belviranlı M, Okudan N. Differential effects of voluntary and forced exercise trainings on spatial learning ability and hippocampal biomarkers in aged female rats. Neurosci Lett 2022; 773:136499. [PMID: 35121056 DOI: 10.1016/j.neulet.2022.136499] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 10/19/2022]
Abstract
This study aimed to compare the effects of voluntary and forced exercise trainings on cognitive functions and to evaluate their relationship with hippocampal synaptic proteins, neurotrophic factors and markers of oxidative damage in aged female rats. Aged female rats were randomly assigned to control, voluntary exercise training and forced exercise training groups. Voluntary or forced exercise trainings were performed for 12 weeks. At the end of the training period, cognitive functions of the animals were assessed with Morris water maze (MWM) test. After the behavioral test, hippocampus tissues were taken for the analysis of synaptophysin, acetylcholinesterase (AChE), brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), malondialdehyde (MDA), protein carbonyl (PC), glutathione (GSH) and superoxide dismutase (SOD). During the MWM test, the number of platform crossings was higher in the voluntary exercise group than in the control group (P < 0.05). In the hippocampus tissue, levels of the synaptophysin, BDNF, NGF and SOD were higher, but MDA levels were lower in the voluntary exercise group than in the control group (P < 0.05). Additionally, hippocampal AChE concentration was higher, but PC levels were lower in the both voluntary and forced exercise groups than in the control group (P < 0.05). In conclusion, voluntary exercise was more effective intervention to improve spatial learning ability in aging process. Increased neurotrophic factors, synaptic proteins, and improved oxidative damage may play a role in these positive effects.
Collapse
Affiliation(s)
- Muaz Belviranlı
- Selçuk University, School of Medicine, Department of Physiology, Konya, Turkey.
| | - Nilsel Okudan
- Selçuk University, School of Medicine, Department of Physiology, Konya, Turkey
| |
Collapse
|
29
|
Cystine/Glutamate Antiporter in Schizophrenia: From Molecular Mechanism to Novel Biomarker and Treatment. Int J Mol Sci 2021; 22:ijms22189718. [PMID: 34575878 PMCID: PMC8466274 DOI: 10.3390/ijms22189718] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 01/04/2023] Open
Abstract
Glutamate, a crucial excitatory neurotransmitter, plays a major role in the modulation of schizophrenia’s pathogenesis. New drug developments for schizophrenia have been prompted by the hypoglutamatergic hypothesis of schizophrenia. The cystine/glutamate antiporter system xc− is related to glutamate-release regulation. Patients with schizophrenia were recently discovered to exhibit downregulation of xc− subunits—the solute carrier (SLC) family 3 member 2 and the SLC family 7 member 11. We searched for relevant studies from 1980, when Bannai and Kitamura first identified the protein subunit system xc− in lung fibroblasts, with the aim of compiling the biological, functional, and pharmacological characteristics of antiporter xc−, which consists of several subunits. Some of them can significantly stimulate the human brain through the glutamate pathway. Initially, extracellular cysteine activates neuronal xc−, causing glutamate efflux. Next, excitatory amino acid transporters enhance the unidirectional transportation of glutamate and sodium. These two biochemical pathways are also crucial to the production of glutathione, a protective agent for neural and glial cells and astrocytes. Investigation of the expression of system xc− genes in the peripheral white blood cells of patients with schizophrenia can facilitate better understanding of the mental disorder and future development of novel biomarkers and treatments for schizophrenia. In addition, the findings further support the hypoglutamatergic hypothesis of schizophrenia.
Collapse
|
30
|
Diabetes and Alzheimer's Disease: Might Mitochondrial Dysfunction Help Deciphering the Common Path? Antioxidants (Basel) 2021; 10:antiox10081257. [PMID: 34439505 PMCID: PMC8389322 DOI: 10.3390/antiox10081257] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
A growing number of clinical and epidemiological studies support the hypothesis of a tight correlation between type 2 diabetes mellitus (T2DM) and the development risk of Alzheimer's disease (AD). Indeed, the proposed definition of Alzheimer's disease as type 3 diabetes (T3D) underlines the key role played by deranged insulin signaling to accumulation of aggregated amyloid beta (Aβ) peptides in the senile plaques of the brain. Metabolic disturbances such as hyperglycemia, peripheral hyperinsulinemia, dysregulated lipid metabolism, and chronic inflammation associated with T2DM are responsible for an inefficient transport of insulin to the brain, producing a neuronal insulin resistance that triggers an enhanced production and deposition of Aβ and concomitantly contributes to impairment in the micro-tubule-associated protein Tau, leading to neural degeneration and cognitive decline. Furthermore, the reduced antioxidant capacity observed in T2DM patients, together with the impairment of cerebral glucose metabolism and the decreased performance of mitochondrial activity, suggests the existence of a relationship between oxidative damage, mitochondrial impairment, and cognitive dysfunction that could further reinforce the common pathophysiology of T2DM and AD. In this review, we discuss the molecular mechanisms by which insulin-signaling dysregulation in T2DM can contribute to the pathogenesis and progression of AD, deepening the analysis of complex mechanisms involved in reactive oxygen species (ROS) production under oxidative stress and their possible influence in AD and T2DM. In addition, the role of current therapies as tools for prevention or treatment of damage induced by oxidative stress in T2DM and AD will be debated.
Collapse
|
31
|
Peixoto Pinheiro B, Adel Y, Knipper M, Müller M, Löwenheim H. Auditory Threshold Variability in the SAMP8 Mouse Model of Age-Related Hearing Loss: Functional Loss and Phenotypic Change Precede Outer Hair Cell Loss. Front Aging Neurosci 2021; 13:708190. [PMID: 34408646 PMCID: PMC8366269 DOI: 10.3389/fnagi.2021.708190] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/09/2021] [Indexed: 11/13/2022] Open
Abstract
Age-related hearing loss (ARHL) is the most common sensory deficit in aging society, which is accompanied by increased speech discrimination difficulties in noisy environments, social isolation, and cognitive decline. The audiometric degree of ARHL is largely correlated with sensory hair cell loss in addition to age-related factors not captured by histopathological analysis of the human cochlea. Previous studies have identified the senescence-accelerated mouse prone strain 8 (SAMP8) as a model for studying ARHL and age-related modifications of the cochlear redox environment. However, the SAMP8 population exhibits a large variability in auditory function decline over age, whose underlying cause remains unknown. In this study, we analyzed auditory function of SAMP8 mice by measuring auditory brainstem response (ABR) thresholds at the age of 6 weeks (juvenile), 12 weeks (young adult), and 24 weeks (adult). Consistent with previous studies, SAMP8 mice exhibit an early progressive, age-related decline of hearing acuity. However, a spatiotemporal cytohistological analysis showed that the significant increase in threshold variability was not concurrently reflected in outer hair cell (OHC) loss observed in the lower and upper quartiles of the ABR threshold distributions over age. This functional loss was found to precede OHC loss suggesting that age-related phenotypic changes may be contributing factors not represented in cytohistological analysis. The expression of potassium channels KCNQ4 (KV7.4), which mediates the current IK,n crucial for the maintenance of OHC membrane potential, and KCNQ1 (KV7.1), which is an essential component in potassium circulation and secretion into the endolymph generating the endocochlear potential, showed differences between these quartiles and age groups. This suggests that phenotypic changes in OHCs or the stria vascularis due to variable oxidative deficiencies in individual mice may be predictors of the observed threshold variability in SAMP8 mice and their progressive ARHL. In future studies, further phenotypic predictors affected by accumulated metabolic challenges over age need to be investigated as potentially underlying causes of ARHL preceding irreversible OHC loss in the SAMP8 mouse model.
Collapse
Affiliation(s)
- Barbara Peixoto Pinheiro
- Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Youssef Adel
- Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Marlies Knipper
- Molecular Physiology of Hearing, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Marcus Müller
- Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Hubert Löwenheim
- Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| |
Collapse
|
32
|
Kaikai NE, Ba-M'hamed S, Ghanima A, Bennis M. Metam sodium exposure during pregnancy and lactation in mice caused behavioral abnormalities and oxidative stress in offspring. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 85:103630. [PMID: 33711515 DOI: 10.1016/j.etap.2021.103630] [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: 09/18/2020] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 06/12/2023]
Abstract
Metam sodium (MS) is a widespread biocide with a broad-spectrum activity. Here, we addressed the behavioral impact of MS by exposing female mice to 50, 100 and 150 mg/kg of MS during both pregnancy and lactation, and evaluated the oxidative stress as a potential mechanism of MS-induced neurotoxicity. The results showed that MS affected fertility and reproduction parameters as well as some aspects of maternal behavior, especially at high doses. In offspring, MS caused a significant delay in the ontogeny of sensorimotor functions. In addition, treated mice exhibited during adulthood an increase of anxiety-like, depression-like behaviors as well as learning and memory impairment. These alterations were accompanied by an increase of the superoxide dismutase activity, and a significant decreased catalase and malondialdehyde activities in specific brain areas. The present work revealed that early exposure to MS induced sensorimotor and behavioral impairments in offspring likely associated with onset of oxidative stress.
Collapse
Affiliation(s)
- Nour-Eddine Kaikai
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Cadi Ayyad University, Faculty of Sciences, Marrakech, Morocco; Research Laboratory for Sustainable Development and Health, Cadi Ayyad University, Faculty of Sciences and Techniques, Marrakech, Morocco
| | - Saadia Ba-M'hamed
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Cadi Ayyad University, Faculty of Sciences, Marrakech, Morocco
| | - Abderrazzak Ghanima
- Research Laboratory for Sustainable Development and Health, Cadi Ayyad University, Faculty of Sciences and Techniques, Marrakech, Morocco.
| | - Mohamed Bennis
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Cadi Ayyad University, Faculty of Sciences, Marrakech, Morocco
| |
Collapse
|
33
|
Hajipour S, Farbood Y, Dianat M, Rashno M, Khorsandi LS, Sarkaki A. Thymoquinone improves cognitive and hippocampal long-term potentiation deficits due to hepatic encephalopathy in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:881-891. [PMID: 34712417 PMCID: PMC8528250 DOI: 10.22038/ijbms.2021.52824.11913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 06/22/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Hepatic encephalopathy (HE) is a neuropsychiatric syndrome that causes brain disturbances. Thymoquinone (TQ) has a wide spectrum of activities such as antioxidant, anti-inflammatory, and anticancer. This study aimed to evaluate the effects of TQ on spatial memory and hippocampal long-term potentiation (LTP) in rats with thioacetamide (TAA)-induced liver injury and hepatic encephalopathy. MATERIALS AND METHODS Adult male Wistar rats were divided into six groups randomly: 1) Control; 2) HE, received TAA (200 mg/kg); 3-5) Treated groups (HE+TQ5, HE+TQ10, and HE+TQ20). TQ (5, 10, and 20 mg/kg) was injected intraperitoneally (IP) for 12 consecutive days from day 18 to 29. Subsequently, spatial memory performance was evaluated by the Morris water maze paradigm and hippocampal LTP was recorded from the dentate gyrus (DG) region. Activity levels of Malondialdehyde (MDA) and superoxide dismutase (SOD) were measured in the hippocampal tissue. RESULTS Data showed that the hippocampal content of MDA was increased while SOD activities were decreased in TAA-induced HE. TQ treatment significantly improved spatial memory and LTP. Moreover, TQ restored the levels of MDA and SOD activities in the hippocampal tissue in HE rats. CONCLUSION Our data confirm that TQ could attenuate cognitive impairment and improve LTP deficit by modulating the oxidative stress parameters in this model of HE, which leads to impairment of spatial cognition and LTP deficit. Thus, these results suggest that TQ may be a promising agent with positive therapeutic effects against liver failure and HE defects.
Collapse
Affiliation(s)
- Somayeh Hajipour
- Persian Gulf Physiology Research Center. Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Yaghoob Farbood
- Persian Gulf Physiology Research Center. Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahin Dianat
- Persian Gulf Physiology Research Center. Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Rashno
- Department of Immunology, Cellular & Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of
| | | | - Alireza Sarkaki
- Persian Gulf Physiology Research Center. Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Anatomical Sciences, Cellular & Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University
| |
Collapse
|
34
|
Oyefeso FA, Muotri AR, Wilson CG, Pecaut MJ. Brain organoids: A promising model to assess oxidative stress-induced central nervous system damage. Dev Neurobiol 2021; 81:653-670. [PMID: 33942547 DOI: 10.1002/dneu.22828] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022]
Abstract
Oxidative stress (OS) is one of the most significant propagators of systemic damage with implications for widespread pathologies such as vascular disease, accelerated aging, degenerative disease, inflammation, and traumatic injury. OS can be induced by numerous factors such as environmental conditions, lifestyle choices, disease states, and genetic susceptibility. It is tied to the accumulation of free radicals, mitochondrial dysfunction, and insufficient antioxidant protection, which leads to cell aging and tissue degeneration over time. Unregulated systemic increase in reactive species, which contain harmful free radicals, can lead to diverse tissue-specific OS responses and disease. Studies of OS in the brain, for example, have demonstrated how this state contributes to neurodegeneration and altered neural plasticity. As the worldwide life expectancy has increased over the last few decades, the prevalence of OS-related diseases resulting from age-associated progressive tissue degeneration. Unfortunately, vital translational research studies designed to identify and target disease biomarkers in human patients have been impeded by many factors (e.g., limited access to human brain tissue for research purposes and poor translation of experimental models). In recent years, stem cell-derived three-dimensional tissue cultures known as "brain organoids" have taken the spotlight as a novel model for studying central nervous system (CNS) diseases. In this review, we discuss the potential of brain organoids to model the responses of human neural cells to OS, noting current and prospective limitations. Overall, brain organoids show promise as an innovative translational model to study CNS susceptibility to OS and elucidate the pathophysiology of the aging brain.
Collapse
Affiliation(s)
- Foluwasomi A Oyefeso
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Alysson R Muotri
- Department of Pediatrics/Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Christopher G Wilson
- Lawrence D. Longo, MD, Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Michael J Pecaut
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
35
|
Molecular Factors Mediating Neural Cell Plasticity Changes in Dementia Brain Diseases. Neural Plast 2021; 2021:8834645. [PMID: 33854544 PMCID: PMC8021472 DOI: 10.1155/2021/8834645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 02/25/2021] [Accepted: 03/16/2021] [Indexed: 11/18/2022] Open
Abstract
Neural plasticity-the ability to alter a neuronal response to environmental stimuli-is an important factor in learning and memory. Short-term synaptic plasticity and long-term synaptic plasticity, including long-term potentiation and long-term depression, are the most-characterized models of learning and memory at the molecular and cellular level. These processes are often disrupted by neurodegeneration-induced dementias. Alzheimer's disease (AD) accounts for 50% of cases of dementia. Vascular dementia (VaD), Parkinson's disease dementia (PDD), dementia with Lewy bodies (DLB), and frontotemporal dementia (FTD) constitute much of the remaining cases. While vascular lesions are the principal cause of VaD, neurodegenerative processes have been established as etiological agents of many dementia diseases. Chief among such processes is the deposition of pathological protein aggregates in vivo including β-amyloid deposition in AD, the formation of neurofibrillary tangles in AD and FTD, and the accumulation of Lewy bodies composed of α-synuclein aggregates in DLB and PDD. The main symptoms of dementia are cognitive decline and memory and learning impairment. Nonetheless, accurate diagnoses of neurodegenerative diseases can be difficult due to overlapping clinical symptoms and the diverse locations of cortical lesions. Still, new neuroimaging and molecular biomarkers have improved clinicians' diagnostic capabilities in the context of dementia and may lead to the development of more effective treatments. Both genetic and environmental factors may lead to the aggregation of pathological proteins and altered levels of cytokines, such that can trigger the formation of proinflammatory immunological phenotypes. This cascade of pathological changes provides fertile ground for the development of neural plasticity disorders and dementias. Available pharmacotherapy and disease-modifying therapies currently in clinical trials may modulate synaptic plasticity to mitigate the effects neuropathological changes have on cognitive function, memory, and learning. In this article, we review the neural plasticity changes seen in common neurodegenerative diseases from pathophysiological and clinical points of view and highlight potential molecular targets of disease-modifying therapies.
Collapse
|
36
|
Daugherty AM. Hypertension-related risk for dementia: A summary review with future directions. Semin Cell Dev Biol 2021; 116:82-89. [PMID: 33722505 DOI: 10.1016/j.semcdb.2021.03.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/01/2021] [Accepted: 03/06/2021] [Indexed: 02/07/2023]
Abstract
Chronic hypertension, or high blood pressure, is the most prevalent vascular risk factor that accelerates cognitive aging and increases risk for Alzheimer's disease and related dementia. Decades of observational and clinical trials have demonstrated that midlife hypertension is associated with greater gray matter atrophy, white matter damage commiserate with demyelination, and functional deficits as compared to normotension over the adult lifespan. Critically, hypertension is a modifiable dementia risk factor: successful blood pressure control with antihypertensive treatment improves outcomes as compared to uncontrolled hypertension, but does not completely negate the risk for dementia. This suggests that hypertension-related risk for neural and cognitive decline in aging cannot be due to elevations in blood pressure alone. This summary review describes three putative pathways for hypertension-related dementia risk: oxidative damage and metabolic dysfunction; systemic inflammation; and autonomic control of heart rate variability. The same processes contribute to pre-clinical hypertension, and therefore hypertension may be an early symptom of an aging nervous system that then exacerbates cumulative and progressive neurodegeneration. Current evidence is reviewed and future directions for research are outlined, including blood biomarkers and novel neuroimaging methods that may be sensitive to test the specific hypotheses.
Collapse
Affiliation(s)
- Ana M Daugherty
- Department of Psychology, Department of Psychiatry and Behavioral Neurosciences, Institute of Gerontology, Wayne State University, 5057 Woodward Ave., Detroit, MI, USA.
| |
Collapse
|
37
|
Macciò A, Oppi S, Madeddu C. COVID-19 and cytokine storm syndrome: can what we know about interleukin-6 in ovarian cancer be applied? J Ovarian Res 2021; 14:28. [PMID: 33550983 PMCID: PMC7868172 DOI: 10.1186/s13048-021-00772-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
Improving early diagnosis along with timely and effective treatment of COVID-19 are urgently needed. However, at present, the mechanisms underlying disease spread and development, defined prognosis, and immune status of patients with COVID-19 remain to be determined. Patients with severe disease state exhibit a hyperinflammatory response associated with cytokine storm syndrome, hypercoagulability, and depressed cell-mediated immunity. These clinical manifestations, sharing similar pathogenesis, have been well-studied in patients with advanced ovarian cancer. The present review suggests treatment approaches for COVID-19 based on strategies used against ovarian cancer, which shares similar immunopathology and associated coagulation disorders.The chronicization of the hyperinflammatory cytokine storm in patients with severe COVID-19 highlights a defective resistance phase that leads to aspecific chronic inflammation, associated with oxidative stress, which impairs specific T-cell response, induces tissue and endothelial damage, and thrombosis associated with systemic effects that lead to severe multi-organ failure and death. These events are similar to those observed in advanced ovarian cancer which share similar pathogenesis mediated primarily by Interleukin-6, which is, as well demonstrated in ovarian cancer, the key cytokine driving the immunopathology, related systemic symptoms, and patient prognosis.Consistent with findings in other disease models with similar immunopathology, such as advanced ovarian cancer, treatment of severe COVID-19 infection should target inflammation, oxidative stress, coagulation disorders, and immunodepression to improve patient outcome. Correctly identifying disease stages, based on available laboratory data, and developing a specific protocol for each phase is essential for effective treatment.
Collapse
Affiliation(s)
- Antonio Macciò
- Department of Gynecologic Oncology, Businco Hospital, "Azienda di Rilievo Nazionale ad Alta Specializzazione G. Brotzu", Via Jenner, 09100, Cagliari, Italy.
| | - Sara Oppi
- Hematology and Transplant Center, Businco Hospital, "Azienda di Rilievo Nazionale ad Alta Specializzazione G. Brotzu", Cagliari, Italy
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
38
|
Age-related hearing loss pertaining to potassium ion channels in the cochlea and auditory pathway. Pflugers Arch 2020; 473:823-840. [PMID: 33336302 PMCID: PMC8076138 DOI: 10.1007/s00424-020-02496-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/27/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022]
Abstract
Age-related hearing loss (ARHL) is the most prevalent sensory deficit in the elderly and constitutes the third highest risk factor for dementia. Lifetime noise exposure, genetic predispositions for degeneration, and metabolic stress are assumed to be the major causes of ARHL. Both noise-induced and hereditary progressive hearing have been linked to decreased cell surface expression and impaired conductance of the potassium ion channel KV7.4 (KCNQ4) in outer hair cells, inspiring future therapies to maintain or prevent the decline of potassium ion channel surface expression to reduce ARHL. In concert with KV7.4 in outer hair cells, KV7.1 (KCNQ1) in the stria vascularis, calcium-activated potassium channels BK (KCNMA1) and SK2 (KCNN2) in hair cells and efferent fiber synapses, and KV3.1 (KCNC1) in the spiral ganglia and ascending auditory circuits share an upregulated expression or subcellular targeting during final differentiation at hearing onset. They also share a distinctive fragility for noise exposure and age-dependent shortfalls in energy supply required for sustained surface expression. Here, we review and discuss the possible contribution of select potassium ion channels in the cochlea and auditory pathway to ARHL. We postulate genes, proteins, or modulators that contribute to sustained ion currents or proper surface expressions of potassium channels under challenging conditions as key for future therapies of ARHL.
Collapse
|
39
|
Kostyuk AI, Panova AS, Kokova AD, Kotova DA, Maltsev DI, Podgorny OV, Belousov VV, Bilan DS. In Vivo Imaging with Genetically Encoded Redox Biosensors. Int J Mol Sci 2020; 21:E8164. [PMID: 33142884 PMCID: PMC7662651 DOI: 10.3390/ijms21218164] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
Redox reactions are of high fundamental and practical interest since they are involved in both normal physiology and the pathogenesis of various diseases. However, this area of research has always been a relatively problematic field in the context of analytical approaches, mostly because of the unstable nature of the compounds that are measured. Genetically encoded sensors allow for the registration of highly reactive molecules in real-time mode and, therefore, they began a new era in redox biology. Their strongest points manifest most brightly in in vivo experiments and pave the way for the non-invasive investigation of biochemical pathways that proceed in organisms from different systematic groups. In the first part of the review, we briefly describe the redox sensors that were used in vivo as well as summarize the model systems to which they were applied. Next, we thoroughly discuss the biological results obtained in these studies in regard to animals, plants, as well as unicellular eukaryotes and prokaryotes. We hope that this work reflects the amazing power of this technology and can serve as a useful guide for biologists and chemists who work in the field of redox processes.
Collapse
Affiliation(s)
- Alexander I. Kostyuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Anastasiya S. Panova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Aleksandra D. Kokova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Daria A. Kotova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Dmitry I. Maltsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Federal Center for Cerebrovascular Pathology and Stroke, 117997 Moscow, Russia
| | - Oleg V. Podgorny
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vsevolod V. Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Federal Center for Cerebrovascular Pathology and Stroke, 117997 Moscow, Russia
- Institute for Cardiovascular Physiology, Georg August University Göttingen, D-37073 Göttingen, Germany
| | - Dmitry S. Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
40
|
Mori Y, Oikawa S, Kurimoto S, Kitamura Y, Tada-Oikawa S, Kobayashi H, Yamashima T, Murata M. Proteomic analysis of the monkey hippocampus for elucidating ischemic resistance. J Clin Biochem Nutr 2020; 67:167-173. [PMID: 33041514 PMCID: PMC7533853 DOI: 10.3164/jcbn.19-78] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/13/2020] [Indexed: 01/25/2023] Open
Abstract
It is well-known that the cornu Ammonis 1 (CA1) sector of hippocampus is vulnerable for the ischemic insult, whereas the dentate gyrus (DG) is resistant. Here, to elucidate its underlying mechanism, alternations of protein oxidation and expression of DG in the monkey hippocampus after ischemia-reperfusion by the proteomic analysis were studied by comparing CA1 data. Oxidative damage to proteins such as protein carbonylation interrupt the protein function. Carbonyl modification of molecular chaperone, heat shock 70 kDa protein 1 (Hsp70.1) was increased remarkably in CA1, but slightly in DG. In addition, expression levels of nicotinamide adenine dinucleotide (NAD)-dependent protein deacetylase sirtuin-2 (SIRT2) was significantly increased in DG after ischemia, but decreased in CA1. Accordingly, it is likely that SIRT2 upregulation and negligible changes of carbonylation of Hsp70.1 exert its neuroprotective effect in DG. On the contrary, carbonylation level of dihydropyrimidinase related protein 2 (DRP-2) and l-lactate dehydrogenase B chain (LDHB) were slightly increased in CA1 as shown previously, but remarkably increased in DG after ischemia. It is considered that DRP-2 and LDHB are specific targets of oxidative stress by ischemia insult and high carbonylation levels of DRP-2 may play an important role in modulating ischemic neuronal death.
Collapse
Affiliation(s)
- Yurie Mori
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Shinji Oikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Shota Kurimoto
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Yuki Kitamura
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan.,College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyama-ku, Nagoya, Aichi 463-8521, Japan
| | - Saeko Tada-Oikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan.,Department of Human Nutrition, School of Life Studies, Sugiyama Jogakuen University, 17-3 Hoshigaoka-motomachi, Chikusa-ku, Nagoya, Aichi 464-8662, Japan
| | - Hatasu Kobayashi
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| | - Tetsumori Yamashima
- Departments of Psychiatry and Neurobiology, Kanazawa University Graduate School of Medical Science, Takakura-machi 13-1, Kanazawa, Ishikawa 920-8641, Japan
| | - Mariko Murata
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie 514-8507, Japan
| |
Collapse
|
41
|
Mohammadali S, Heshami N, Komaki A, Tayebinia H, Abbasi Oshaghi E, Karimi J, Hashemnia M, Khodadadi I. Dill tablet and Ocimum basilicum aqueous extract: Promising therapeutic agents for improving cognitive deficit in hypercholesterolemic rats. J Food Biochem 2020; 44:e13485. [PMID: 33015851 DOI: 10.1111/jfbc.13485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/14/2020] [Accepted: 09/06/2020] [Indexed: 01/13/2023]
Abstract
High-cholesterol diet (HCD) is correlated with Alzheimer's disease (AD) and impairment of memory. This study investigated beneficial therapeutic effects of Dill tablet and Ocimum basilicum (Basil) aqueous extract on hypercholesterolemia-induced cognitive deficits and oxidative stress in hippocampus tissues of rats. Hippocampal Aβ(1-42) level was measured. The gene expression levels of superoxide dismutase and inducible-nitric oxide synthase were determined in hippocampus. Cognitive functions were examined and oxidative status was evaluated in serum and hippocampus. Phytochemical properties and in vitro antioxidant activity of Basil extract were assessed. HCD significantly increased serum cholesterol, induced deposition of Aβ plaque, altered hippocampus morphology, and impaired memory function, whereas receiving Basil extract or Dill tablet increased antioxidant potency in serum and hippocampus and normalized HCD-induced deleterious effects. Basil extract and Dill tablet may exhibit their beneficial effects in AD by lowering serum cholesterol and evoking antioxidant system in the brain. PRACTICAL APPLICATIONS: Dill tablet and Basil aqueous extract lowered serum cholesterol in hypercholesterolemic animal models, therefore, they can be used as hypocholesterolemic agents. These edible herbs significantly retarded deposition of Aβ plaque and normalized hippocampal morphology, thus, they favorably protected hippocampus tissue from deleterious effects-induced by hypercholesterolemia. Dill tablet and Basil aqueous extract also corrected oxide-redox balance and normalized HCD-induced oxidative stress to some extent and significantly improved impairments in learning and memory suggesting that these medicinal plants can be considered as surrogate therapeutic agents for the synthetic medicines in the treatment of AD and in postponement of its complications.
Collapse
Affiliation(s)
- Soheila Mohammadali
- Faculty of Medicine, Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Neda Heshami
- Faculty of Medicine, Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Heidar Tayebinia
- Faculty of Medicine, Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ebrahim Abbasi Oshaghi
- Faculty of Medicine, Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jamshid Karimi
- Faculty of Medicine, Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Hashemnia
- Faculty of Veterinary Medicine, Department of Pathobiology, Razi University, Kermanshah, Iran
| | - Iraj Khodadadi
- Faculty of Medicine, Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran.,Research Center for Nutrition Health, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
42
|
Flores NP, Bona NP, Luduvico KP, Cardoso JDS, Soares MSP, Gamaro GD, Spanevello RM, Lencina CL, Gazal M, Stefanello FM. Eugenia uniflora fruit extract exerts neuroprotective effect on chronic unpredictable stress-induced behavioral and neurochemical changes. J Food Biochem 2020; 44:e13442. [PMID: 32803896 DOI: 10.1111/jfbc.13442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 01/01/2023]
Abstract
The aim of the current study was to evaluate the effect of chronic administration of Eugenia uniflora fruit extract on behavioral parameters, oxidative stress markers, and acetylcholinesterase activity in an animal model of depression, which was induced by chronic unpredictable stress (CUS). Mice were divided into six groups as follows: control/vehicle (water), control/fluoxetine (20 mg/kg), control/extract (200 mg/kg), CUS/vehicle, CUS/fluoxetine (20 mg/kg), and CUS/extract (200 mg/kg). Animals of the CUS group were exposed to a series of stressors for a period of 21 days. Vehicle, fluoxetine, and hydroalcoholic extract were administered daily by gavage. Results showed that E. uniflora treatment: (a) prevented the depressant-like effect induced by CUS; (b) regulated the activity of acetylcholinesterase; (c) reduced oxidative damage to lipids and reactive oxygen species production, in the prefrontal cortex and hippocampus; and (d) prevented the reduction of glutathione peroxidase in the hippocampus of animals subjected to CUS protocol. Taken together, our findings suggested that E. uniflora extract exerts a neuroprotective effect by preventing oxidative damage and decreasing CUS-induced acetylcholinesterase activity, thus, ameliorating depressive-type behavior. PRACTICAL APPLICATIONS: E. uniflora fruit extract revealed an antidepressant-like effect and prevented the oxidative damage as well as cholinergic alterations caused by chronic stress in mice. Therefore, we believe that the results obtained in this study can be used to develop an alternative therapy for the management of depressive disorders.
Collapse
Affiliation(s)
- Natália Porto Flores
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Natália Pontes Bona
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Karina Pereira Luduvico
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Juliane de Souza Cardoso
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Mayara Sandrielly Pereira Soares
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Giovana Duzzo Gamaro
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Roselia Maria Spanevello
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Claiton Leoneti Lencina
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Marta Gazal
- Programa de Biologia Celular e Molecular-Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Francieli Moro Stefanello
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| |
Collapse
|
43
|
Sharma VK, Singh TG. Insulin resistance and bioenergetic manifestations: Targets and approaches in Alzheimer's disease. Life Sci 2020; 262:118401. [PMID: 32926928 DOI: 10.1016/j.lfs.2020.118401] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/04/2020] [Accepted: 09/05/2020] [Indexed: 12/15/2022]
Abstract
AIM Insulin has a well-established role in cognition, neuronal detoxification and synaptic plasticity. Insulin transduction affect neurotransmitter functions, influence bioenergetics and regulate neuronal survival through regulating glucose energy metabolism and downward pathways. METHODS A systematic literature review of PubMed, Medline, Bentham, Scopus and EMBASE (Elsevier) databases was carried out with the help of the keywords like "Alzheimer's disease; Hypometabolism; Oxidative stress; energy failure in AD, Insulin; Insulin resistance; Bioenergetics" till June 2020. The review was conducted using the above keywords to collect the latest articles and to understand the nature of the extensive work carried out on insulin resistance and bioenergetic manifestations in Alzheimer's disease. KEY FINDINGS The article sheds light on insulin resistance mediated hypometabolic state on pathological progression of AD. The disrupted insulin signaling has pathological outcome in form of disturbed glucose homeostasis, altered bioenergetic state which increases build-up of senile plaques (Aβ), neurofibrillary tangles (τ), decline in transportation of glucose and activation of inflammatory pathways. The mechanistic link of insulin resistant state with therapeutically explorable potential transduction pathways is the focus of the reviewed work. SIGNIFICANCE The present work opines that the mechanism by which the insulin resistance mediates dysregulation of bioenergetics and progresses to neurodegenerative state holds the tangible potential to succeed in the development of novel dementia therapies. Further, hypometabolic complications and altered insulin signaling may be explored as a mechanistic relation between bioenergetic deficits and AD.
Collapse
Affiliation(s)
- Vivek Kumar Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; Govt. College of Pharmacy, Rohru, District Shimla, Himachal Pradesh 171207, India
| | | |
Collapse
|
44
|
Ardeshiri MR, Beheshti SA, Beheshti F, Hosseini M, Akbari E. Curcumin modulates long term potentiation in lipopolysaccharide-treated rats. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
45
|
Umukoro S, Ben-Azu B, Ajayi AM, Adebesin A, Emokpae O. Cymbopogon citratus aqueous leaf extract attenuates neurobehavioral and biochemical changes induced by social defeat stress in mice. CHINESE HERBAL MEDICINES 2020; 12:303-309. [PMID: 36119005 PMCID: PMC9476801 DOI: 10.1016/j.chmed.2020.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/02/2019] [Accepted: 01/12/2020] [Indexed: 11/20/2022] Open
Abstract
Objective Psychosocial stress has been implicated in the genesis of psychiatric disorders such as memory deficits, depression, anxiety and addiction. Aqueous leaf extract of Cymbopogon citratus (CYC) otherwise known as lemongrass tea has antidepressant, anxiolytic and anti-amnesic effects in rodents. This study was designed to evaluate if C. citratus could reverse the neurobehavioral and biochemical derangements induced by social defeat stress (SDS) in the resident/intruder paradigm. Methods Intruder male mice were divided into five groups (n = 7): group 1 received saline (10 mL/kg, p.o.; non-stress control), group 2 also received saline (10 mL/kg, p.o.; SDS control) while groups 3–5 had C. citratus (50, 100 and 200 mg/kg, p.o.) daily for 14 d. The SDS was carried out 30 min after each treatment from day 7 to day 14 by exposing each intruder mouse in groups 2–5 to a 10 min confrontation in the home cage of an aggressive resident counterpart. The neurobehavioral features (spontaneous motor activity-SMA, anxiety, memory, social avoidance and depression were then evaluated. The concentrations of nitrite, malondialdehyde and glutathione as well as acetylcholinesterase activity in the brain tissues were also determined. Results C. citratus (50, 100 and 200 mg/kg) attenuated hypolocomotion, heightened anxiety, depressive-like symptom, memory deficit and social avoidance induced by SDS. The altered levels of oxidative stress and acetyl-cholinesterase in SDS-mice were positively modulated by C. citratus. Conclusion The results of this study suggest that C. citratus might mitigate psychosocial stress-induced neurologic diseases in susceptible individuals.
Collapse
Affiliation(s)
- Solomon Umukoro
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan 200284, Nigeria
- Corresponding author.
| | - Benneth Ben-Azu
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan 200284, Nigeria
- Department of Pharmacology, Faculty of Basic Medical Sciences, PAMO University of Medical Sciences, Port Harcourt 500272, Nigeria
| | - Abayomi M. Ajayi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan 200284, Nigeria
| | - Adaeze Adebesin
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan 200284, Nigeria
- Department of Biochemistry, University of Africa, Toru-Orua 561, Nigeria
| | - Osagie Emokpae
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan 200284, Nigeria
| |
Collapse
|
46
|
Santa-Helena E, Seus N, Castro M. Effect of pristine fullerene on acquisition, consolidation and retrieval memory in wistar rats. Comp Biochem Physiol C Toxicol Pharmacol 2020; 232:108740. [PMID: 32169415 DOI: 10.1016/j.cbpc.2020.108740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/29/2020] [Accepted: 03/06/2020] [Indexed: 11/29/2022]
Abstract
The present study evaluated the effect of fullerene (C60) under in vitro conditions, in hippocampus homogenates from rats and on the induction of behavioral disabilities. Exposure to in vitro C60 led to an increase in the concentration of reactive oxygen species (ROS) and lipid peroxidation (LPO) of hippocampus treated with of fullerene and suspension. These results indicate that the oxidative stress caused by the exposure to C60 was in part related to an absence of an antioxidant response. In this sense, one-trial inhibitory avoidance task were performed and results showed that fullerene at 0.2 and 0.45 μm impaired the acquisition and consolidation of short and long-term memory. Further, enzymatic analysis in rat hippocampus were not significantly different, however, there was an increase in the content of LPO and ROS produced by fullerene. Overall, the results indicates that fullerene possess neurotoxic properties that impairs behavior and promotes oxidative stress.
Collapse
Affiliation(s)
- Eduarda Santa-Helena
- Universidade Federal do Rio Grande, Instituto de Ciências Biológicas, Programa de Pós-Graduação em Ciências Fisiológicas-PPGCF-FURG, Av. Itália km 8, 96203-900, Rio Grande, RS
| | - Natália Seus
- Universidade Federal do Rio Grande, Instituto de Ciências Biológicas, Programa de Pós-Graduação em Ciências Fisiológicas-PPGCF-FURG, Av. Itália km 8, 96203-900, Rio Grande, RS
| | - Micheli Castro
- Universidade Federal do Rio Grande, Instituto de Ciências Biológicas, Programa de Pós-Graduação em Ciências Fisiológicas-PPGCF-FURG, Av. Itália km 8, 96203-900, Rio Grande, RS.
| |
Collapse
|
47
|
Preparation of sea cucumber (Stichopus variegates) peptide fraction with desired organoleptic property and its anti-aging activity in fruit flies and D-galactose-induced aging mice. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103954] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
48
|
Muñoz P, Ardiles ÁO, Pérez-Espinosa B, Núñez-Espinosa C, Paula-Lima A, González-Billault C, Espinosa-Parrilla Y. Redox modifications in synaptic components as biomarkers of cognitive status, in brain aging and disease. Mech Ageing Dev 2020; 189:111250. [PMID: 32433996 DOI: 10.1016/j.mad.2020.111250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/05/2020] [Accepted: 04/10/2020] [Indexed: 02/06/2023]
Abstract
Aging is a natural process that includes several changes that gradually make organisms degenerate and die. Harman's theory proposes that aging is a consequence of the progressive accumulation of oxidative modifications mediated by reactive oxygen/nitrogen species, which plays an essential role in the development and progression of many neurodegenerative diseases. This review will focus on how abnormal redox modifications induced by age impair the functionality of neuronal redox-sensitive proteins involved in axonal elongation and guidance, synaptic plasticity, and intercellular communication. We will discuss post-transcriptional regulation of gene expression by microRNAs as a mechanism that controls the neuronal redox state. Finally, we will discuss how some brain-permeant antioxidants from the diet have a beneficial effect on cognition. Taken together, the evidence revised here indicates that oxidative-driven modifications of specific proteins and changes in microRNA expression may be useful biomarkers for aging and neurodegenerative diseases. Also, some specific antioxidant therapies have undoubtedly beneficial neuroprotective effects when administered in the correct doses, in the ideal formulation combination, and during the appropriate therapeutic window. The use of some antioxidants is, therefore, still poorly explored for the treatment of neurodegenerative diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- Pablo Muñoz
- Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile; Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile; Biomedical Research Center, Universidad de Valparaíso, Valparaíso, Chile; Thematic Task Force on Healthy Aging, CUECH Research Network.
| | - Álvaro O Ardiles
- Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile; Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile; Thematic Task Force on Healthy Aging, CUECH Research Network; Interdisciplinary Center of Neuroscience of Valparaíso, Universidad de Valparaíso, Valparaíso, Chile; Interdisciplinary Center for Health Studies, Universidad de Valparaíso, Valparaíso, Chile
| | - Boris Pérez-Espinosa
- Thematic Task Force on Healthy Aging, CUECH Research Network; Laboratorio biología de la Reproduccion, Departamento Biomédico, Facultad Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Cristian Núñez-Espinosa
- Thematic Task Force on Healthy Aging, CUECH Research Network; School of Medicine, Universidad de Magallanes, Punta Arenas, Chile
| | - Andrea Paula-Lima
- Thematic Task Force on Healthy Aging, CUECH Research Network; Institute for Research in Dental Sciences, Faculty of Dentistry; Universidad de Chile, Santiago, Chile; Biomedical Neuroscience Institute (BNI) and Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Christian González-Billault
- Thematic Task Force on Healthy Aging, CUECH Research Network; Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; FONDAP Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, USA.
| | - Yolanda Espinosa-Parrilla
- Thematic Task Force on Healthy Aging, CUECH Research Network; School of Medicine, Universidad de Magallanes, Punta Arenas, Chile; Laboratory of Molecular Medicine - LMM, Center for Education, Healthcare and Investigation - CADI, University of Magallanes, Punta Arenas, Chile.
| |
Collapse
|
49
|
Findley CA, Bartke A, Hascup KN, Hascup ER. Amyloid Beta-Related Alterations to Glutamate Signaling Dynamics During Alzheimer's Disease Progression. ASN Neuro 2020; 11:1759091419855541. [PMID: 31213067 PMCID: PMC6582288 DOI: 10.1177/1759091419855541] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) ranks sixth on the Centers for Disease Control and Prevention Top 10 Leading Causes of Death list for 2016, and the Alzheimer’s Association attributes 60% to 80% of dementia cases as AD related. AD pathology hallmarks include accumulation of senile plaques and neurofibrillary tangles; however, evidence supports that soluble amyloid beta (Aβ), rather than insoluble plaques, may instigate synaptic failure. Soluble Aβ accumulation results in depression of long-term potentiation leading to cognitive deficits commonly characterized in AD. The mechanisms through which Aβ incites cognitive decline have been extensively explored, with a growing body of evidence pointing to modulation of the glutamatergic system. The period of glutamatergic hypoactivation observed alongside long-term potentiation depression and cognitive deficits in later disease stages may be the consequence of a preceding period of increased glutamatergic activity. This review will explore the Aβ-related changes to the tripartite glutamate synapse resulting in altered cell signaling throughout disease progression, ultimately culminating in oxidative stress, synaptic dysfunction, and neuronal loss.
Collapse
Affiliation(s)
- Caleigh A Findley
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- 3 Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N Hascup
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.,4 Department of Molecular Biology, Microbiology & Biochemistry, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R Hascup
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
50
|
La Rosa P, Petrillo S, Bertini ES, Piemonte F. Oxidative Stress in DNA Repeat Expansion Disorders: A Focus on NRF2 Signaling Involvement. Biomolecules 2020; 10:biom10050702. [PMID: 32369911 PMCID: PMC7277112 DOI: 10.3390/biom10050702] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
DNA repeat expansion disorders are a group of neuromuscular and neurodegenerative diseases that arise from the inheritance of long tracts of nucleotide repetitions, located in the regulatory region, introns, or inside the coding sequence of a gene. Although loss of protein expression and/or the gain of function of its transcribed mRNA or translated product represent the major pathogenic effect of these pathologies, mitochondrial dysfunction and imbalance in redox homeostasis are reported as common features in these disorders, deeply affecting their severity and progression. In this review, we examine the role that the redox imbalance plays in the pathological mechanisms of DNA expansion disorders and the recent advances on antioxidant treatments, particularly focusing on the expression and the activity of the transcription factor NRF2, the main cellular regulator of the antioxidant response.
Collapse
|