101
|
Ren H, Tang L, Yuan Z, Liu Y, Zhou X, Xiao X, Wu X, Chen W, Chen Y, Wang H, Xue Q, Xu X. Combined administration of catalpol, puerarin, gastrodin, and borneol modulates the Tlr4/Myd88/NF-κB signaling pathway and alleviates microglia inflammation in Alzheimer's disease. Front Pharmacol 2024; 15:1492237. [PMID: 39545064 PMCID: PMC11560463 DOI: 10.3389/fphar.2024.1492237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder affecting millions of people worldwide, with no effective treatment currently available. In recent decades, various traditional Chinese medicines (TCMs) and their active ingredients have shown the potential to attenuate the pathogenesis of AD in cellular and animal models. However, the effects of TCM formulas, which are typically administered in practice, have been less studied. This study aims to investigate the therapeutic effects of several formulas consisting of 4 components herbal components: catalpol, puerarin, gastrodin, and borneol, on streptozotocin (STZ)-induced AD models in cells and rats. The new object recognition (NOR), elevated plus maze (EMP), and Morris water maze (MWM) tests were used to evaluate the cognitive functions of rats. Golgi staining, Haematoxylin and Eosin (HE) staining, and Nissl staining analyses were employed assess the physiology of hippocampal tissues. Gene expression profiles were analyzed used transcriptomics and reverse transcription quantitative polymerase chain reaction analysis, while protein expression levels were determined using immunoblotting, immunohistochemical, and immunofluorescence. The production of cytokines was evaluated with enzyme-linked immunosorbent assay. The results demonstrated that the combined administration of these 4 components (CPGB) had superior mitigating effects on AD cell model, as evidenced by the reduced pro-inflammatory cytokine production and decreased deposition of Aβ protein. Further in vivo and in vitro experiments confirmed that varying doses of CPGB formula effectively ameliorated STZ-induced cognitive deficits, as shown by NOR, MWM, and EMP tests, as well as pathological changes in hippocampal tissues and a 3-dimensional brain neurovascular unit (3D-NVU) model, including decreased deposition of Aβ protein and formation of plaques. Transcriptome sequencing and analysis identified 35 genes with significantly altered expression levels due to STZ and CPGB treatment in hippocampal tissues, which were enriched in the Tlr4/Myd88/NF-κB signaling pathway. Interference with this pathway significantly influenced the therapeutic effects of CPGB in the 3D-NVU model. Collectively, these findings suggest that the combined administration of catalpol, puerarin, gastrodin, and borneol offers superior therapeutic effects on AD by modulating the Tlr4/Myd88/NF-κB signaling pathway. This study strengthens the theoretical foundation for using TCMs to treat AD, proving new insights and references for alleviating and treating AD.
Collapse
Affiliation(s)
- Huijing Ren
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- Tongren Polytechnic College, Tongren, Guizhou, China
| | - Ling Tang
- Shapingba District People’s Hospital of Chongqing, Chongqing, China
| | - Zhiying Yuan
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Yang Liu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xuejiao Zhou
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xiao Xiao
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xingyu Wu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Weihai Chen
- Faculty of Psychology, Southwest University, Chongqing, China
| | - Yi Chen
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Hongjin Wang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Qiang Xue
- Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Xiaoyu Xu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- Southwest University Hospital, Chongqing, China
- Chongqing Key Laboratory of New Drug Screening form Traditional Chinese Medicine, Chongqing, China
- Key Disciplines of Traditional Chinese Medicine of Chongqing City, Rehabilitation Medicine of Southwest University, Chongqing, China
| |
Collapse
|
102
|
Liu B, Dong K, Chen X, Dong H, Zhao Y, Wang X, Sun Z, Xie F, Qian L. Inhibition of Glycolysis Alleviates Chronic Unpredictable Mild Stress Induced Neuroinflammation and Depression-like Behavior. Brain Sci 2024; 14:1098. [PMID: 39595861 PMCID: PMC11591872 DOI: 10.3390/brainsci14111098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Growing evidence suggests that glucose metabolism plays a crucial role in activated immune cells, significantly contributing to the occurrence and development of neuroinflammation and depression-like behaviors. Chronic stress has been reported to induce microglia activation and disturbances in glucose metabolism in the hippocampus. AIMS This study aims to investigate how chronic stress-mediated glycolysis promotes neuroinflammation and to assess the therapeutic potential of the glycolysis inhibitor, 2-deoxy-D-glucose (2-DG), in a model of chronic stress-induced neuroinflammation and depression-like behavior. METHODS In in vitro studies, we first explored the effects of 2-DG on the inflammatory response of microglia cells. The results showed that corticosterone (Cort) induced reactive oxygen species (ROS) production, increased glycolysis, and promoted the release of inflammatory mediators. However, these effects were reversed by intervention with 2-DG. Subsequently, we examined changes in depression-like behavior and hippocampal glycolysis in mice during chronic stress. The results indicated that chronic stress led to prolonged escape latency in the Morris water maze, increased platform-crossing frequency, reduced sucrose preference index, and extended immobility time in the forced swim test, all of which are indicative of depression-like behavior in mice. Additionally, we found that the expression of the key glycolytic enzyme hexokinase 2 (HK2) was upregulated in the hippocampus of stressed mice, along with an increased release of inflammatory factors. Further in vivo experiments investigated the effects of 2-DG on glycolysis and pro-inflammatory mediator production, as well as the therapeutic effects of 2-DG on chronic stress-induced depression-like behavior in mice. The results showed that 2-DG alleviated chronic stress-induced depression-like behaviors, such as improving escape latency and platform-crossing frequency in the Morris water maze, and increasing the time spent in the center of the open field. Additionally, 2-DG intervention reduced the level of glycolysis in the hippocampus and decreased the release of pro-inflammatory mediators. CONCLUSIONS These findings suggest that 2-DG can mitigate neuroinflammation and depressive behaviors by inhibiting glycolysis and inflammatory responses. Overall, our results highlight the potential of 2-DG as a therapeutic agent for alleviating chronic stress-induced neuroinflammation through the regulation of glycolysis.
Collapse
Affiliation(s)
- Bing Liu
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Ke Dong
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
- School of Medicine, South China University of Technology, Guangzhou 511442, China
| | - Xiaobing Chen
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Huafeng Dong
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Yun Zhao
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Xue Wang
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Zhaowei Sun
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Fang Xie
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| | - Lingjia Qian
- Beijing Institute of Basic Medical Sciences, #27 Taiping Road, Haidian, Beijing 100039, China; (B.L.); (K.D.); (X.C.); (H.D.); (Y.Z.); (X.W.); (Z.S.); (F.X.)
| |
Collapse
|
103
|
Martínez-Drudis L, Bérard M, Musiol D, Rivest S, Oueslati A. Pharmacological inhibition of PLK2 kinase activity mitigates cognitive decline but aggravates APP pathology in a sex-dependent manner in APP/PS1 mouse model of Alzheimer's disease. Heliyon 2024; 10:e39571. [PMID: 39498012 PMCID: PMC11532864 DOI: 10.1016/j.heliyon.2024.e39571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Converging evidence from clinical and experimental studies suggest the potential significance of Polo-like kinase 2 (PLK2) in regulating the phosphorylation and toxicity of the Alzheimer's disease (AD)-related protein, amyloid precursor protein (APP). These findings have prompted various experimental trials aimed at inhibiting PLK2 kinase activity in different transgenic mouse models of AD. While positive impacts on cognitive decline were reported in these studies, the cellular effects remained controversial. In the present study, we sought to assess the cognitive and cellular consequences of chronic PLK2 inhibitor treatment in the APP/PS1 transgenic mouse model of AD. First, we confirmed that inhibiting PLK2 prevented cognitive decline in a sex-dependent manner, particularly by enhancing working memory in male APP/PS1 mice. Surprisingly, cellular analysis revealed that treatment with PLK2 inhibitor increased the load of amyloid plaques and elevated levels of soluble amyloid β (Aβ) 40 and Aβ42 in the cortex, as well as insoluble Aβ42 in the hippocampus of female mice, without affecting APP pathology in males. These results underscore the potential of PLK2 inhibition to mitigate cognitive symptoms in males. However, paradoxically, it intensifies amyloid pathology in females by enhancing APP amyloidogenic processing, creating a controversial aspect to its therapeutic impact. Overall, these data highlight the sex-dependent nature of the effects of PLK2 inhibition, which may also be influenced by the genetic background of the transgenic mouse model utilized.
Collapse
Affiliation(s)
- Laura Martínez-Drudis
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Morgan Bérard
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Dylan Musiol
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Serge Rivest
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Abid Oueslati
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| |
Collapse
|
104
|
Wu Y, Gao R, Huang Q, Huang C, Wang L, Lin L, He G, Wu K, Liu X, Liu X, Liu L. Lactate supplementation after hypoglycemia alleviates cognitive dysfunction induced by recurrent non-severe hypoglycemia in diabetic mice. Exp Neurol 2024; 383:115037. [PMID: 39481512 DOI: 10.1016/j.expneurol.2024.115037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Recurrent non-severe hypoglycemia (RH) in diabetes is an independent risk factor for cognitive dysfunction. However, the mechanisms and potential therapeutic strategies remain poorly understood. In this study, we aimed to elucidate the mechanisms underlying RH-induced diabetic cognitive impairment. We investigated the effects of RH on lactate metabolism and cognitive function in male C57BL/6 J diabetic mice. After RH, diabetic mice showed decreased brain lactate and adenosine triphosphate levels, decreased expression of lactate transporter proteins MCT1 and MCT4, increased neuroapoptosis, and decreased astrocyte glycolysis in vitro. This was accompanied by increased neuronal mitochondrial reactive oxygen species levels, decreased mitochondrial COX IV activity, impaired mitochondrial morphology and function, impaired synaptic morphology, and decreased expression of synaptic plasticity proteins. Intraperitoneal lactic acid injection improved lactate transport restored neuronal mitochondrial morphology and function, upregulated synaptic plasticity proteins brain-derived neurotrophic factor and early growth response 1, enhanced synaptic ultrastructure, and ultimately improved cognitive dysfunction following RH in diabetic mice. These findings provide insights into the prevention and treatment of cognitive dysfunction in patients with diabetes mellitus caused by RH.
Collapse
Affiliation(s)
- Yubin Wu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Ruonan Gao
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Qintao Huang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Cuihua Huang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Lijing Wang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Lu Lin
- 900th Hospital of the Joint Logistic Support Force, Fuzhou 350000, China
| | - Guanlian He
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Kejun Wu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xiaoying Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xiaohong Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Libin Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| |
Collapse
|
105
|
Shao N, Ding Z, Liu F, Zhang X, Wang X, Hu S, Ye S, Wang T, Si W, Cai B. Huang-Pu-Tong-Qiao Formula Alleviates Hippocampal Neuron Damage by Inhibiting NLRP3 Inflammasome-mediated Pyroptosis in Alzheimer's Disease. Mol Neurobiol 2024:10.1007/s12035-024-04547-0. [PMID: 39466576 DOI: 10.1007/s12035-024-04547-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024]
Abstract
Huang-Pu-Tong-Qiao (HPTQ), a Traditional Chinese Medicine formula, has achieved remarkable efficacy in clinically treating Alzheimer's disease (AD). Pyroptosis refers to the inflammatory necrosis of cells, which contributes to AD pathological progression. However, it is unclear whether the therapeutic effect of HPTQ on AD is related to reducing pyroptosis. In this study, the network pharmacology analysis was used to predict the molecular mechanism of HPTQ in treating AD and validated our hypothesis through mice and cell experiments. APP/PS1 transgenic mice and Aβ25-35-injured HT22 cells were used as AD models in vivo and in vitro. The pharmacological effects and mechanisms of HPTQ on AD were evaluated by Morris water maze, Y-maze, transmission electron microscope, immunofluorescence, Hoechst/PI staining, western blot, and ELISA. Network pharmacology reveals the correlation between the therapeutic effect of HPTQ on AD and the NOD-like receptor signaling pathway. In APP/PS1 mice, HPTQ reduced the escape latency and maintained cell membrane integrity. In HT22 cells, 15% HPTQ-medicated serum and 10 µM MCC950 increased cell viability and decreased PI positive rate compared with the Model group. In addition, HPTQ treatment in AD animal and cell models reduced the protein expressions of NLRP3, ASC, cleaved caspase-1, GSDMD, GSDMD-N, IL-1β, and IL-18. The experimental results of MCC950 specifically inhibiting the NLRP3 expression suggested that HPTQ might reduce neuronal pyroptosis by reducing NLRP3 inflammasome. Network pharmacology and experimental validation suggested that HPTQ alleviated NLRP3 inflammasome-mediated neuronal pyroptosis in AD, which could provide valuable candidate drugs for AD clinical treatment.
Collapse
Affiliation(s)
- Nan Shao
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Zhixian Ding
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Fei Liu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Xiaoyan Zhang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Xiaojuan Wang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Shenglin Hu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Key Laboratory of Xin'an Medicine, Anhui University of Chinese Medicine, Ministry of Education, Hefei, 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| | - Shu Ye
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Key Laboratory of Xin'an Medicine, Anhui University of Chinese Medicine, Ministry of Education, Hefei, 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| | - Tingting Wang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Key Laboratory of Xin'an Medicine, Anhui University of Chinese Medicine, Ministry of Education, Hefei, 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| | - Wenwen Si
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China.
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China.
- Key Laboratory of Xin'an Medicine, Anhui University of Chinese Medicine, Ministry of Education, Hefei, 230012, China.
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China.
| | - Biao Cai
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China.
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China.
- Key Laboratory of Xin'an Medicine, Anhui University of Chinese Medicine, Ministry of Education, Hefei, 230012, China.
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China.
| |
Collapse
|
106
|
Liu PP, Liu XH, Ren MJ, Liu XT, Shi XQ, Li ML, Li SA, Yang Y, Wang DD, Wu Y, Yin FX, Guo YH, Yang RZ, Cheng M, Xin YJ, Kang JS, Huang B, Ren KD. Neuronal cathepsin S increases neuroinflammation and causes cognitive decline via CX3CL1-CX3CR1 axis and JAK2-STAT3 pathway in aging and Alzheimer's disease. Aging Cell 2024:e14393. [PMID: 39453382 DOI: 10.1111/acel.14393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Aging is an intricate process involving interactions among multiple factors, which is one of the main risks for chronic diseases, including Alzheimer's disease (AD). As a member of cysteine protease, cathepsin S (CTSS) has been implicated in inflammation across various diseases. Here, we investigated the role of neuronal CTSS in aging and AD started by examining CTSS expression in hippocampus neurons of aging mice and identified a significant increase, which was negatively correlated with recognition abilities. Concurrently, we observed an elevation of CTSS concentration in the serum of elderly people. Transcriptome and fluorescence-activated cell sorting (FACS) results revealed that CTSS overexpression in neurons aggravated brain inflammatory milieu with microglia activation to M1 pro-inflammatory phenotype, activation of chemokine C-X3-C-motif ligand 1 (CX3CL1)-chemokine C-X3-C-motif receptor 1 (CX3CR1) axis and janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) pathway. As CX3CL1 is secreted by neurons and acts on the CX3CR1 in microglia, our results revealed for the first time the role of neuron CTSS in neuron-microglia "crosstalk." Besides, we observed elevated CTSS expression in multiple brain regions of AD patients, including the hippocampus. Utilizing CTSS selective inhibitor, LY3000328, rescued AD-related pathological features in APP/PS1 mice. We further noticed that neuronal CTSS overexpression increased cathepsin B (CTSB) activity, but decreased cathepsin L (CTSL) activity in microglia. Overall, we provide evidence that CTSS can be used as an aging biomarker and plays regulatory roles through modulating neuroinflammation and recognition in aging and AD process.
Collapse
Affiliation(s)
- Pei-Pei Liu
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao-Hui Liu
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ming-Jing Ren
- Department of Nephropathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao-Tong Liu
- Department of Clinical Laboratory, The First Hospital of Yongnian District, Hebei, China
| | - Xiao-Qing Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ming-Li Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shu-Ang Li
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dian-Dian Wang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yue Wu
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fan-Xiang Yin
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan-Hong Guo
- Department of Nephropathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Run-Zhou Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Meng Cheng
- Henan Branch, Bank of China, Zhengzhou, Henan, China
| | - Yong-Juan Xin
- Department of Child and Adolescent Health, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Jian-Sheng Kang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bing Huang
- Pain and Related Disease Research Laboratory, Shantou University Medical College, Shantou, Guangdong, China
| | - Kai-Di Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University4, Zhengzhou, Henan, China
| |
Collapse
|
107
|
Pandey KK, Mehta K, Kaur B, Dhar P. Curcumin alleviates arsenic trioxide-induced neural damage in the murine striatal region. Psychopharmacology (Berl) 2024:10.1007/s00213-024-06700-y. [PMID: 39443330 DOI: 10.1007/s00213-024-06700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024]
Abstract
RATIONALE Arsenic-induced neurotoxicity, with dose-dependent effects, is well-documented in rodents. Curcumin (CUR), a cost-effective plant polyphenol, shows neuroprotective effects by modulating oxidative stress, apoptosis, and neurochemistry. This study evaluates curcumin's neuroprotective potential against arsenic trioxide (As2O3) in the mouse striatal region. METHODS Healthy adult male mice were chronically administered with varying concentrations of As2O3 (2, 4 and 8 mg/kg bw) alone and along with CUR (100 mg/kg bw) orally for 45 days. Towards the end of the experimental period, the animals were subjected to behavioural paradigms including open field task, novel object recognition, rota-rod, and Morris water maze. Striatal tissues were freshly collected from the animals on day 46 for biochemical analyses (MDA, GPx, and GSH). Additionally, perfusion-fixed brains were processed for morphological observations. RESULTS Behavioural study showed an apparent decrease in certain cognitive functions (learning and memory) and locomotor activity in mice exposed to As2O3 compared to controls. Simultaneous treatment of As2O3 (2, 4 and 8 mg/kg bw) and curcumin (100 mg/kg bw) alleviated the As-induced locomotor and cognitive deficits. As2O3 alone exposure also exhibited a significant increase in oxidative stress marker (MDA) and a decrease in antioxidant enzyme levels (GPx, GSH). Morphological alterations were noted in mice subjected to elevated doses of As2O3 (4 and 8 mg/kg bw). However, these changes were reversed in mice who received As2O3 + CUR co-treatment. CONCLUSIONS Collectively, our findings indicate that curcumin offers neuroprotection to the striatal region against As2O3-induced behavioral deficits, as well as biochemical and morphological alterations.
Collapse
Affiliation(s)
- Kamlesh Kumar Pandey
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Kamakshi Mehta
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, 110029, India.
- Department of Ophthalmology, University of Pittsburgh School of Medicine, UPMC Vision Institute, Pittsburgh, PA, 15219, USA.
| | - Balpreet Kaur
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Pushpa Dhar
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
108
|
Salazar-García M, Villavicencio-Guzmán L, Revilla-Monsalve C, Patiño-Morales CC, Jaime-Cruz R, Ramírez-Fuentes TC, Corona JC. Harmful Effects on the Hippocampal Morpho-Histology and on Learning and Memory in the Offspring of Rats with Streptozotocin-Induced Diabetes. Int J Mol Sci 2024; 25:11335. [PMID: 39518888 PMCID: PMC11546933 DOI: 10.3390/ijms252111335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Learning alterations in the child population may be linked to gestational diabetes as a causal factor, though this remains an open and highly controversial question. In that sense, it has been reported that maternal hyperglycemia generates a threatening condition that affects hippocampal development in offspring. The pyramidal cells of the CA3 subfield, a key structure in learning and memory processes, are particularly important in cognitive deficiencies. We evaluate the effect of the hyperglycemic intrauterine environment on hippocampal histomorphometry in offspring, correlating it with spatial learning and memory, as well as the morphology of dendrites and spines in 30-day-old pups (P30). The maternal hyperglycemia affected the body weight, height, and brain size of fetuses at 21 days of gestation (F21), newborn pups (P0) and P30 pups from diabetic rats, which were smaller compared to the control group. Consequently, this resulted in a decrease in hippocampal size, lower neuronal density and cytoarchitectural disorganization in the CA3 region of the hippocampus in the offspring at the three ages studied. The behavioral tests performed showed a direct relationship between morpho-histological alterations and deficiencies in learning and memory, as well as alterations in the morphology of the dendrites and spines. Therefore, knowing the harmful effects caused by gestational diabetes can be of great help to establish therapeutic and educational strategies that can help to improve learning and memory in children.
Collapse
Affiliation(s)
- Marcela Salazar-García
- Laboratorio de Investigación en Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (L.V.-G.); (C.C.P.-M.)
| | - Laura Villavicencio-Guzmán
- Laboratorio de Investigación en Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (L.V.-G.); (C.C.P.-M.)
| | - Cristina Revilla-Monsalve
- Unidad de Investigación en Enfermedades Metabólicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06725, Mexico;
| | - Carlos César Patiño-Morales
- Laboratorio de Investigación en Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (L.V.-G.); (C.C.P.-M.)
| | - Ricardo Jaime-Cruz
- Departamento de Ciencias de la Salud, Universidad Tecnológica de México-UNITEC México-Campus Sur, Mexico City 09810, Mexico;
| | - Tania Cristina Ramírez-Fuentes
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Juan Carlos Corona
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| |
Collapse
|
109
|
Abdi SMY, Al-Bakri SSM, Nordin N. Insights on the Characteristics and Therapeutic Potential of Mesenchymal Stem Cell-derived Exosomes for Mitigation of Alzheimer's Disease's Pathogenicity: A Systematic Review. Cell Biochem Biophys 2024:10.1007/s12013-024-01598-x. [PMID: 39436580 DOI: 10.1007/s12013-024-01598-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 10/23/2024]
Abstract
Alzheimer's disease (AD) remains a progressive neurodegenerative disease with no cure. Treatment of AD relies on administering drugs that only subside the symptoms. In recent studies, mesenchymal stem cell (MSC)-exosomes have been marked to possess therapeutic potential for treating AD. This study aims to systematically review and analyse findings that focus on the isolation, characterisation, and sources of MSC-derived exosomes used to unravel the therapeutic potential of these exosomes targeting AD using in vitro and in vivo models. It is hypothesised that MSC-exosomes exhibit high therapeutic potential for AD treatment by exerting various modes of action. PubMed, Scopus, and Medline were used to find relevant published works from January 2016 until December 2020, using assigned keywords including "Alzheimer's disease", "secretome", and "exosomes". Only research articles meeting the predefined inclusion/exclusion criteria were selected and analysed. The risk of bias was assessed using the Office of Health Assessment and Translation tool (OHAT). A total of 17 eligible in vivo and in vitro studies were included in this review. Bone marrow-derived stem cells (BMSCs) were the most used source for exosome isolation, even though studies on exosomes from adipose-derived stem cells (ADSCs) and human umbilical cord stem cells (HUCSCs) provide more information on the characteristics. When the risk of bias was assessed, the studies presented various levels of biases. Notably, the in vitro and in vivo studies revealed neuroprotective properties of MSC-exosomes through different modes of action to alleviate AD pathology. Our review discovered that most MSC exosomes could degrade Aβ plaques, enhance neurogenesis, extenuate neuroinflammatory response through microglial activation, regulate apoptosis and reduce oxidative stress. Delivery of exosomal micro-RNAs was also found to reduce neuroinflammation. Findings from this review provided convincing systematic evidence highlighting the therapeutic properties of MSC-derived exosomes as a prospective source for cell-free (acellular) therapy in treating AD.
Collapse
Affiliation(s)
- Sarah Mohammed Yousuf Abdi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Siti Sarah Mustaffa Al-Bakri
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Norshariza Nordin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
- Malaysian Research Institute on Ageing (MyAgeing™), Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
- Genetics & Regenerative Medicine (ReGEN) Research Group, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
| |
Collapse
|
110
|
Islam T, Torigoe M, Tanimoto Y, Okamoto H. Adult zebrafish can learn Morris water maze-like tasks in a two-dimensional virtual reality system. CELL REPORTS METHODS 2024; 4:100863. [PMID: 39317191 PMCID: PMC11573742 DOI: 10.1016/j.crmeth.2024.100863] [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: 03/26/2024] [Revised: 06/11/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024]
Abstract
Virtual reality (VR) has emerged as a powerful tool for investigating neural mechanisms of decision-making, spatial cognition, and navigation. In many head-fixed VRs for rodents, animals locomote on spherical treadmills that provide rotation information in two axes to calculate two-dimensional (2D) movement. On the other hand, zebrafish in a submerged head-fixed VR can move their tail to enable movement in 2D VR environment. This motivated us to create a VR system for adult zebrafish to enable 2D movement consisting of forward translation and rotations calculated from tail movement. Besides presenting the VR system, we show that zebrafish can learn a virtual Morris water maze-like (VMWM) task in which finding an invisible safe zone was necessary for the zebrafish to avoid an aversive periodic mild electric shock. Results show high potential for our VR system to be combined with optical imaging for future studies to investigate spatial learning and navigation.
Collapse
Affiliation(s)
- Tanvir Islam
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan.
| | - Makio Torigoe
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | - Yuki Tanimoto
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan; School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatu-cho, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Hitoshi Okamoto
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan; School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatu-cho, Shinjuku-ku, Tokyo 169-8555, Japan; Institute of Neuropsychiatry, 91 Benten-cho, Shinjuku-ku, Tokyo 162-0851, Japan.
| |
Collapse
|
111
|
Zhang Z, Li R, Zhou Y, Huang S, Hou Y, Pei G. Dietary Flavonoid Chrysin Functions as a Dual Modulator to Attenuate Amyloid-β and Tau Pathology in the Models of Alzheimer's Disease. Mol Neurobiol 2024:10.1007/s12035-024-04557-y. [PMID: 39432184 DOI: 10.1007/s12035-024-04557-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
Growing evidence indicates that healthy diets are associated with a slower progression of Alzheimer's disease (AD). Flavonoids are among the most abundant natural products in diets beneficial to AD, such as the Mediterranean diet. However, the effect and mechanism of these dietary flavonoids on AD remains incompletely understood. Here, we found that a representative dietary natural flavonoid, chrysin (Chr), significantly ameliorated cognitive impairment and AD pathology in APP/PS1 mice. Furthermore, mechanistic studies showed that Chr significantly reduced the levels of amyloid-β (Aβ) and phosphorylated tau (p-tau), along with dual inhibitory activity against β-site amyloid precursor protein cleaving enzyme 1 (BACE1) and glycogen synthase kinase 3β (GSK3β). Moreover, the effect of Chr was further confirmed by EW233, a structural analog of Chr that exhibited an improved pharmacokinetic profile. To further verify the role of Chr and EW233, we utilized our previously established chimeric human cerebral organoid (chCO) model for AD, in which astrogenesis was promoted to mimic the neuron-astrocyte ratio in human brain tissue, and similar dual inhibition of Aβ and p-tau was also observed. Altogether, our study not only reveals the molecular mechanisms through which dietary flavonoids, such as Chr, mitigate AD pathology, but also suggests that identifying a specific constituent that mimics some of the benefits of these healthy diets could serve as a promising approach to discover new treatments for AD.
Collapse
Affiliation(s)
- Zhen Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Rongyao Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yue Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shichao Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yujun Hou
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Gang Pei
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Biomedicine, The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
112
|
Wang X, Wang Q, Wang X, Zhao H, Zhao C, Jiao Y, Shi H, Chen C, Chen H, Wang P, Song T. Early intervention using long-term rhythmic pulsed magnetic stimulation alleviates cognitive decline in a 5xFAD mouse model of Alzheimer's disease. Exp Neurol 2024; 383:115002. [PMID: 39419435 DOI: 10.1016/j.expneurol.2024.115002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/02/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent form of dementia, but no effective therapeutic strategy is available to date. Rhythmic magnetic stimulation is an attractive means of neuron modulation that could be beneficial for restoring learning and memory abilities. OBJECTIVE To assess the effect of a compound pulsed rhythmic magnetic field (cPMF) on cognition during AD progression and to explore the appropriate cPMF intervention period. METHODS Female 5xFAD mice aged 10 weeks and 18 weeks were exposed to cPMF with a carrier frequency of 40 Hz, repeated at 5 Hz for 1 h/d for 8 consecutive weeks. The Morris water maze (MWM) test was used for cognitive behavioral assessment. Furthermore, changes in molecular pathology within the brain were detected using immunofluorescence staining and real-time PCR. RESULTS 10-week-old AD mice treated with cPMF explored the target quadrant more frequently than sham-exposed AD mice in MWM test, exhibiting improved learning and memory abilities. Additionally, cPMF exposure alleviated Aβ plaque deposition and astrogliosis in the AD brain. Moreover, neurotrophic factor fibroblast growth factor 1 (FGF1) in the AD brain was upregulated by cPMF treatment. However, in 18-week-old AD mice treated with cPMF, cognitive performance and Fgf1 gene expression were not significantly improved, although Aβ plaque deposition and astrogliosis were alleviated. CONCLUSION Early intervention via long-term rhythmic cPMF stimulation may alleviate the histopathological features and enhance neuroprotective gene Fgf1 expression, thereby improving the cognitive performance of 5xFAD mice, which should provide promising insight for the clinical treatment of patients with AD.
Collapse
Affiliation(s)
- Xue Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Qingmeng Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Xuting Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Haoyu Zhao
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Chuncheng Zhao
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yangkun Jiao
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Hongkai Shi
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Changyou Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Haitao Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Pingping Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| | - Tao Song
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of the Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
113
|
Shintani T, Yanai S, Kanasaki A, Iida T, Endo S. Long-term d-allose administration ameliorates age-related cognitive impairment and loss of bone strength in male mice. Exp Gerontol 2024; 196:112555. [PMID: 39179160 DOI: 10.1016/j.exger.2024.112555] [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: 05/22/2024] [Revised: 07/12/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Age-related physical and cognitive decline may be ameliorated by consuming functional foods. d-Allose, reported to have multiple health benefits, may temper aging phenotypes, particularly brain function. We investigated whether d-allose supplementation improves cognitive function. A standard battery of behavioral tests was administered to 18-month-old male mice after consuming diet containing 3 % d-allose for 6 months. Following a wire-hanging test, an open-field test, Morris water maze, fear-conditioning, and an analgesia test were sequentially performed. Bone density and strength were assessed afterwards. Possible mechanism(s) under-lying memory changes in hippocampus were also examined with a DNA microarray. d-Allose failed to influence muscle strength, locomotor activity and anxiety, fear memory, or pain sensitivity. However, d-allose improved hippocampus-dependent spatial learning and memory, and it may contribute to increase bone strength. d-Allose also changed the expression of some genes in hippocampus involved in cognitive functions. Long-term d-allose supplementation appears to modestly change aging phenotypes and improve spatial memory.
Collapse
Affiliation(s)
- Tomoya Shintani
- Research and Development, Matsutani Chemical Industry Co., Ltd., 5-3 Kita-Itami, Itami City, Hyogo 664-8508, Japan
| | - Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan; Animal Facility, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Akane Kanasaki
- Research and Development, Matsutani Chemical Industry Co., Ltd., 5-3 Kita-Itami, Itami City, Hyogo 664-8508, Japan.
| | - Tetsuo Iida
- Research and Development, Matsutani Chemical Industry Co., Ltd., 5-3 Kita-Itami, Itami City, Hyogo 664-8508, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
114
|
Khan JZ, Zainab SR, Rehman MU, Abid M, Mazhar MU, Shah FA, Tipu MK. Chronic stress intensify PTZ-induced seizures by triggering neuroinflammation and oxidative stress. Biochem Biophys Res Commun 2024; 729:150333. [PMID: 38991397 DOI: 10.1016/j.bbrc.2024.150333] [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: 05/07/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Epilepsy is a paroxysmal abnormal hypersynchronous electrical discharge characterized by recurrent seizures. It affects more than 50 million people worldwide. Stress is the leading cause of neurodegeneration and can produce seizures that may lead to or aggravate epilepsy. Inflammation plays a vital role in epilepsy by modulating oxidative stress, and levels of neuroinflammatory cytokines including NF-κB, TNF-α, and IL-1β. METHODS Stress-induced changes in behavior were evaluated in mice by employing behavioral assessment tests such as an elevated plus maze, light-dark box, open field test, tail suspension test, Y-maze, novel object recognition test, and Morris water maze in pentylenetetrazole (PTZ) kindled mice. Behavioral changes in all these paradigms including seizure score, latency, and frequency showed an increase in symptoms in PTZ (35 mg/kg) induced seizures in stressed mice (RS-PTZ) as compared to PTZ, Stress, and normal animals. RESULTS The Enzyme-linked immunosorbent assay (ELISA) results confirmed increased in serum cortisol levels. Histological examinations showed neurodegenerative changes in the hippocampus and cortex regions. The spectrophotometric evaluation showed an increase in oxidative stress by decreasing antioxidant production i.e. reduced glutathione, glutathione -s- transferase, and catalase (CAT), and increasing oxidant levels such as maloaldehyde and nitric oxide. Immunohistochemistry results showed increased expression of NF-κB, TNF-α, and IL-1β in the cortex and hippocampus of mice brains. CONCLUSIONS Results from the study conclude that stress increases the likelihood of eliciting an epileptic attack by increasing the level of reactive oxygen species and neuroinflammation.
Collapse
Affiliation(s)
- Jehan Zeb Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Syeda Rida Zainab
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | | | - Muhammad Abid
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Muhammad Usama Mazhar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Fawad Ali Shah
- Department of Pharmacology and Toxicology, College of Pharmacy Prince Sattam Bin Abdulaziz University, Al-Kharj, 16278, Saudi Arabia.
| | - Muhammad Khalid Tipu
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
115
|
Swift NA, Yang Q, Jester HM, Zhou X, Manuel A, Kemp BE, Steinberg GR, Ma T. Suppression of neuronal AMPKβ2 isoform impairs recognition memory and synaptic plasticity. Neurobiol Dis 2024; 201:106664. [PMID: 39278510 PMCID: PMC11539201 DOI: 10.1016/j.nbd.2024.106664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024] Open
Abstract
AMP-activated protein kinase (AMPK) is an αβγ heterotrimer protein kinase that functions as a molecular sensor to maintain energy homeostasis. Accumulating evidence suggests a role of AMPK signaling in the regulation of synaptic plasticity and cognitive function; however, isoform-specific roles of AMPK in the central nervous system (CNS) remain elusive. Regulation of the AMPK activities has focused on the manipulation of the α or γ subunit. Meanwhile, accumulating evidence indicates that the β subunit is critical for sensing nutrients such as fatty acids and glycogen to control AMPK activity. Here, we generated transgenic mice with conditional suppression of either AMPKβ1 or β2 in neurons and characterized potential isoform-specific roles of AMPKβ in cognitive function and underlying mechanisms. We found that AMPKβ2 (but not β1) suppression resulted in impaired recognition memory, reduced hippocampal synaptic plasticity, and altered structure of hippocampal postsynaptic densities and dendritic spines. Our study implicates a role for the AMPKβ2 isoform in the regulation of synaptic and cognitive function.
Collapse
Affiliation(s)
- Nathaniel A Swift
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Qian Yang
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Hannah M Jester
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Xueyan Zhou
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Adam Manuel
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Bruce E Kemp
- St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia; Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne 3000, VIC, Australia
| | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Tao Ma
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA; Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| |
Collapse
|
116
|
Zhong Z, Liu J, Luo Y, Wu M, Qiu F, Zhao H, Liu Y, Wang Y, Long H, Zhao L, Wang Y, Han Y, Meng P. Jujuboside A Regulates Calcium Homeostasis and Structural Plasticity to Alleviate Depression-Like Behavior via Shh Signaling in Immature Neurons. Drug Des Devel Ther 2024; 18:4565-4584. [PMID: 39416424 PMCID: PMC11482263 DOI: 10.2147/dddt.s479055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Background Depression, a leading cause of disability worldwide, is characterized by dysfunction of immature neurons, resulting in dysregulated calcium homeostasis and impaired structural plasticity. Jujuboside A (JuA), a biologically active compound derived from Semen Ziziphi Spinosae, has demonstrated anti-anxiety and anti-insomnia properties. Recent studies suggest that JuA may be a promising antidepressant, but its underlying mechanisms remain unclear. Methods Sprague-Dawley rats were subjected to chronic unpredictable mild stress (CUMS) to induce a depression model. JuA (12.5 mg/kg, 25 mg/kg, 50 mg/kg) was administered orally for 4 weeks. Emotional and cognitive function were assessed. Monoamine neurotransmitter levels were measured using enzyme-linked immunosorbent assay (ELISA). The number of immature neurons and calcium homeostasis were evaluated by immunofluorescence. Western blotting and immunofluorescence were employed to detect the expression of Sonic hedgehog (Shh) signaling proteins. Additionally, lentiviral vector expressing Shh shRNA (LV-Shh-RNAi) were infused intracerebrally to investigate the role of Shh in JuA's antidepressant effects. Results JuA significantly ameliorated depressive-like behavior and cognitive dysfunction in CUMS rats, increased monoamine neurotransmitter levels in serum and hippocampal tissue, reduced the number of BrdU/DCX (bromodeoxyuridine/doublecortin)-positive immature neurons, and attenuated calcium ion (Ca2+) concentration and Ca2+/calmodulin-dependent protein kinase II (CaMKII) levels in immature neurons. JuA also markedly elevated synaptic density and prominence complexity, upregulated Shh, Gli family zinc finger 1 and 2 (Gli1/2), synaptophysin (Syn) and postsynaptic density protein-95 (PSD-95) expression in the ventral dentate gyrus (vDG). However, knockdown of Shh in the vDG counteracted JuA's therapeutic effects. Conclusion These findings collectively suggest that JuA improves depressive-like behavior in CUMS rats by modulating calcium homeostasis and synaptic structural plasticity in immature neurons through the Shh signaling pathway.
Collapse
Affiliation(s)
- Ziyan Zhong
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Jian Liu
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Yan Luo
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Mei Wu
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Feng Qiu
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Hongqing Zhao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Yang Liu
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Yajing Wang
- Office of Science & Technology, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Hongping Long
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Lei Zhao
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Yuhong Wang
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Yuanshan Han
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Pan Meng
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| |
Collapse
|
117
|
Xiong X, Hou J, Zheng Y, Jiang T, Zhao X, Cai J, Huang J, He H, Xu J, Qian S, Lu Y, Wang X, Wang W, Ye Q, Zhou S, Lian M, Xiao J, Song W, Xie C. NAD +-boosting agent nicotinamide mononucleotide potently improves mitochondria stress response in Alzheimer's disease via ATF4-dependent mitochondrial UPR. Cell Death Dis 2024; 15:744. [PMID: 39394148 PMCID: PMC11470026 DOI: 10.1038/s41419-024-07062-1] [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: 04/04/2024] [Revised: 09/01/2024] [Accepted: 09/06/2024] [Indexed: 10/13/2024]
Abstract
Extensive studies indicate that mitochondria dysfunction is pivotal for Alzheimer's disease (AD) pathogenesis; while cumulative evidence suggests that increased mitochondrial stress response (MSR) may mitigate neurodegeneration in AD, explorations to develop a MSR-targeted therapeutic strategy against AD are scarce. We combined cell biology, molecular biology, and pharmacological approaches to unravel a novel molecular pathway by which NAD+-boosting agent nicotinamide mononucleotide (NMN) regulates MSR in AD models. Here, we report dyshomeostasis plasma UPRmt-mitophagy-mediated MSR profiles in AD patient samples. NMN restores NAD+ metabolic profiles and improves MSR through the ATF4-dependent UPRmt pathway in AD-related cross-species models. At the organismal level, NAD+ repletion with NMN supplementation ameliorates mitochondrial proteotoxicity, decreases hippocampal synaptic disruption, decreases neuronal loss, and brain atrophy in mice model of AD. Remarkably, omics features of the hippocampus with NMN show that NMN leads to transcriptional changes of genes and proteins involved in MSR characteristics, principally within the astrocyte unit rather than microglia and oligodendrocytes. In brief, our work provides evidence that MSR has an active role in the pathogenesis of AD, as reducing mitochondrial homeostasis via atf4 depletion in AD mice aggravates the hallmarks of the disease; conversely, bolstering mitochondrial proteostasis by NMN decreases protein aggregation, restores memory performance, and delays disease progression, ultimately translating to increased healthspan.
Collapse
Affiliation(s)
- Xi Xiong
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jialong Hou
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Zheng
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Jiang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuemiao Zhao
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinlai Cai
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiani Huang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haijun He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiaxue Xu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuangjie Qian
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yao Lu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Neurology, Yuhuan City People's Hospital, Taizhou, China
| | - XinShi Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenwen Wang
- The Center of Traditional Chinese Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qianqian Ye
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuoting Zhou
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengjia Lian
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, China
| | - Jian Xiao
- Oujiang Laboratory, Wenzhou, Zhejiang, China.
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China.
| | - Weihong Song
- Oujiang Laboratory, Wenzhou, Zhejiang, China.
- Key Laboratory Of Alzheimer's Disease Of Zhejiang Province, Institute Of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Chenglong Xie
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Oujiang Laboratory, Wenzhou, Zhejiang, China.
- Key Laboratory Of Alzheimer's Disease Of Zhejiang Province, Institute Of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Department of Geriatrics, Geriatric Medical Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
118
|
Zaky DA, Mehny KA, Abdelrahman SS, El-Yamany MF, Kamel AS. Flibanserin conquers murine depressive pseudodementia by amending HPA axis, maladaptive inflammation and AKT/GSK/STAT/BDNF trajectory: Center-staging of the serotonergic/adrenergic circuitry. Eur J Pharmacol 2024; 980:176869. [PMID: 39117265 DOI: 10.1016/j.ejphar.2024.176869] [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: 03/06/2024] [Revised: 07/10/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Depressive pseudodementia (DPD) is a debilitating cognitive dysfunction that accompanies major and/or frequent depressive attacks. DPD has gained significant research attention owing to its negative effects on the patients' quality of life and productivity. This study tested the procognitive potential of Flibanserin (FBN), the serotonin (5HT) receptor modulator, against propranolol (PRP), as β/5HT1A receptors blocker. Serving this purpose, female Wistar Albino rats were subjected to chronic unpredictable stress (CUS) and subsequently treated with FBN only (3 mg/kg/day, p.o), PRP only (10 mg/kg/day, p.o), or PRP followed by FBN, using the same doses. FBN ameliorated the behavioral/cognitive alterations and calmed the hypothalamic-pituitary-adrenal (HPA) axis storm by reducing the levels of stress-related hormones, viz, corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), corticosterone (CORT) parallel to epinephrine (EPI) hyperstimulation. The maladaptive inflammatory response, comprising of interleukin (IL)-1β/6, and tumor necrosis factor (TNF)-α, was consequently blunted. This was contemporaneous to the partial restoration of the protein kinase-B (AKT)/glycogen synthase kinase (GSK)3β/signal transducer and activator of transcription (STAT)-3 survival trajectory and the reinstatement of the levels of brain derived neurotrophic factor (BDNF). Microscopically, FBN repaired the hippocampal architecture and lessened CD68/GFAP immunoreactivity. Pre-administration of PRP partially abolished FBN effect along the estimated parameters, except for 5HT2A receptor expression and epinephrine level, to prove 5HT1A receptor as a fulcrum initiator of the investigated pathway, while its sole administration worsened the underlying condition. Ultimately, these findings highlight the immense procognitive potential of FBN, offering a new paradigm for halting DPD advancement via synchronizing adrenergic/serotonergic circuitry.
Collapse
Affiliation(s)
- Doaa A Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo, 11562, Egypt.
| | | | - Sahar S Abdelrahman
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo, 11562, Egypt
| | - Ahmed S Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo, 11562, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| |
Collapse
|
119
|
Mohammadkhanizadeh A, Hosseini Y, Nikbakht F, Parvizi M, Khodabandehloo F. Evaluating the potential effects of apigenin on memory, anxiety, and social interaction amelioration after social isolation stress. Int J Dev Neurosci 2024. [PMID: 39367711 DOI: 10.1002/jdn.10380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/11/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024] Open
Abstract
Vigorous research confirmed the anti-inflammatory, antioxidant, and antidementia effects of apigenin (Api). The present study evaluated the beneficial impacts of Api administration on behaviour, brain-derived neurotrophic factor (BDNF), Interleukin 6 (IL-6), oxidative stress, and inflammation induced by social isolation (SI) stress in rats. For this purpose, rats underwent a 28-day SI period followed by a 4-week oral Api treatment (50 mg/kg/day, PO). On Day 56, behaviour tests were performed, including an elevated plus maze (EPM), Morris water maze (MWM), and three-chamber social tests. The oxidative stress markers, IL-6, and BDNF levels were measured in the hippocampus. Our results showed that SI stress caused an increase in anxiety and a decrease in spatial memory, sociability, and social preference index. In addition, SI stress increased hippocampal levels of IL-6 and malondialdehyde (MDA) content, whereas it reduced the hippocampal BDNF level and superoxide dismutase (SOD) activities. Our study indicated that Api attenuates anxiety and causes improvements in spatial memory and social interaction. These desirable effects of Api might be related to amelioration in the BDNF level, IL-6, and oxidative stress biomarkers in the hippocampus.
Collapse
Affiliation(s)
- Ali Mohammadkhanizadeh
- Behavioural and Cognitive Science Research Centre, AJA University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Yasaman Hosseini
- Behavioural and Cognitive Science Research Centre, AJA University of Medical Sciences, Tehran, Iran
| | - Farnaz Nikbakht
- Cellular and Molecular Research Center and Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Parvizi
- Behavioural and Cognitive Science Research Centre, AJA University of Medical Sciences, Tehran, Iran
- Department of Physiology, faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khodabandehloo
- Department of Genetic and Advanced Medicine Technology, faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
120
|
Badawi GA, Shokr MM, Elshazly SM, Zaki HF, Mohamed AF. "Sigma-1 receptor modulation by clemastine highlights its repurposing as neuroprotective agent against seizures and cognitive deficits in PTZ-kindled rats". Eur J Pharmacol 2024; 980:176851. [PMID: 39084454 DOI: 10.1016/j.ejphar.2024.176851] [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: 04/30/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Epilepsy is a neurological disorder characterized by recurrent spontaneous seizures alongside other neurological comorbidities. Cognitive impairment is the most frequent comorbidity secondary to progressive neurologic changes in epilepsy. Sigma 1 receptors (σ1 receptors) are involved in the neuroprotection and pathophysiology of both conditions and targeting these receptors may have the potential to modulate both seizures and comorbidities. The current research demonstrated the effect of clemastine (10 mg/kg, P.O.), a non-selective σ1 receptor agonist, on pentylenetetrazol (PTZ) (35 mg/kg, i.p., every 48 h for 14 doses)-kindling rats by acting on σ1 receptors through its anti-inflammatory/antioxidant capacity. Clemastine and phenytoin (30 mg/kg, P.O.) or their combination were given once daily. Clemastine treatment showed a significant effect on neurochemical, behavioural, and histopathological analyses through modulation of σ1 receptors. It protected the kindling animals from seizures and attenuated their cognitive impairment in the Morris water maze test by reversing the PTZ hippocampal neuroinflammation/oxidative stress state through a significant increase in inositol-requiring enzyme 1 (IRE1), x-box binding protein 1 (XBP1), along with a reduction of total reactive oxygen species (TROS) and amyloid beta protein (Aβ). The involvement of σ1 receptors in the protective effects of clemastine was confirmed by their abrogation when utilizing NE-100, a selective σ1 receptor antagonist. In light of our findings, modulating σ1 receptors emerges as a compelling therapeutic strategy for epilepsy and its associated cognitive impairments. The significant neuroprotective effects observed with clemastine underscore the potential of σ1 receptor-targeted treatments to address both the primary symptoms and comorbidities of neurological disorders.
Collapse
Affiliation(s)
- Ghada A Badawi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt
| | - Mustafa M Shokr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt.
| | - Shimaa M Elshazly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Faculty of Pharmacy, King Salman International University (KSIU), South Sinai, 46612, Egypt
| |
Collapse
|
121
|
Long J, Ren Z, Duan Y, Tao W, Li X, Li S, Li K, Huang Q, Chen J, Yang M, Li Y, Luo X, Liu D. Empagliflozin rescues lifespan and liver senescence in naturally aged mice. GeroScience 2024; 46:4969-4986. [PMID: 38922380 PMCID: PMC11336130 DOI: 10.1007/s11357-024-01250-9] [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: 01/26/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Empagliflozin is currently known to decrease blood glucose levels, delay renal failure, and reduce the risk of cardiovascular death and all-cause mortality in patients with type 2 diabetes with cardiovascular disease. However, the effects of empagliflozin on the lifespan and health of naturally aged organisms are unclear. This study was designed to investigate the impacts and potential mechanisms of empagliflozin on lifespan and liver senescence in naturally aged mice. Our study revealed that empagliflozin improved survival and health in naturally aged mice. Empagliflozin extended the median survival of male mice by 5.9%. Meanwhile, empagliflozin improved learning memory and motor balance, decreased body weight, and downregulated the hepatic protein expression of P21, P16, α-SMA, and COL1A1. Empagliflozin modulates the structure of the intestinal flora, increasing the relative abundance of Lachnospiraceae, Ruminococcaceae, Lactobacillus, Blautia, and Muribaculaceae and decreasing the relative abundance of Erysipelotrichaceae, Turicibacter, and Dubosiella in naturally aged mice. Further exploration discovered that empagliflozin increased the concentration of SCFAs, decreased the levels of the inflammatory factors TNF-α, IL-6, and CXCL9, and regulated the PI3K/AKT/P21 and AMPK/SIRT1/NF-κB pathways, which may represent the underlying mechanisms involved in these beneficial hepatic effects. Taken together, the above results indicated that empagliflozin intervention could be considered a potential strategy for extending lifespan and slowing liver senescence in naturally aged mice.
Collapse
Affiliation(s)
- Jiangchuan Long
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Ziyu Ren
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Yaqian Duan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Wei Tao
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400010, China
| | - Xi Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, 400010, China
| | - Shengbing Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Ke Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Qixuan Huang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Jie Chen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Mengliu Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Yang Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Xie Luo
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Dongfang Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
- Chongqing Clinical Research Center for Geriatrics and Gerontology, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
122
|
Su X, Li Q, Yang M, Zhang W, Liu X, Ba Y, Deng Q, Zhang Y, Han L, Huang H. Resveratrol protects against a high-fat diet-induced neuroinflammation by suppressing mitochondrial fission via targeting SIRT1/PGC-1α. Exp Neurol 2024; 380:114899. [PMID: 39059737 DOI: 10.1016/j.expneurol.2024.114899] [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: 01/10/2024] [Revised: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Various health issues have emerged due to consuming high-fat diets (HFD), particularly the detrimental impact they have on mitochondrial dynamics and subsequet cognition functions. Specially, mitochondrial fission can serve as an upstream signal in the regulation of cortical inflammation and neural pyroptosis. Our study was designed to verify the existence of neuroinflammation in the pathogenesis of HFD-induced cognitive dysfunction and demonstrated that resveratrol (RSV) attenuated neural deficits via regulation of cortical mitochondrial fission. A total of 50 male Sprague Dawley rats were randomly divided into five groups: control (Cont, 26 weeks on normal rodent diet); high-fat diet (HFD); dietary adjustments (HFD + ND); resveratrol intervention (HFD + R); joint intervention (HFD + ND + R) for 26 weeks. The spatial learning and memory function, spine density, NLRP3 inflammasome associated protein, mRNA and protein expression involved in mitochondrial dynamics and SIRT1/PGC-1α signaling pathway in brain were measured. Furthermore, reactive oxygen species (ROS) accumulation and resultant mitochondrial membrane potential (MMP) alteration in PC12 cells exposed to palmitic acid (PA) or Drp1 inhibitor (Mdivi-1) were detected to reflect mitochondrial function. The findings suggested that prolonged treatment of RSV improved cognitive deficits and neuronal damage induced by HFD, potentially attributed to activation of the SIRT1/PGC-1α axis. We further indicated that the activation of the NLRP3 inflammasome in PA (200 μM) treated PC12 cells could be inhibited by Mdivi-1. More importantly, Mdivi-1 (10 μM) reduced intracellular ROS levels and enhanced MMP by reversing Drp1-mediated aberrant mitochondrial fission. To summarize, those results clearly indicated that a HFD inhibited the SIRT1/PGC-1α pathway, which contributed to an imbalance in mitochondrial dynamics and the onset of NLRP3-mediated pyroptosis. This effect was mitigated by the RSV possibly through triggering the SIRT1/PGC-1α axis, prevented aberrant mitochondrial fission and thus inhibited the activation of the NLRP3 inflammatory pathway.
Collapse
Affiliation(s)
- Xiao Su
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Qiong Li
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Mingzhi Yang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Wenhui Zhang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Xiaoxue Liu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Yue Ba
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Qihong Deng
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Yu Zhang
- State Key Laboratory of Microbial Technology, Qingdao, Shandong 266000, China; Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong 266000, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250100, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong 266000, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250100, China
| | - Hui Huang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China.
| |
Collapse
|
123
|
Sasaki K, Becker J, Ong J, Ciaghi S, Guldin LS, Savastano S, Fukumitsu S, Kuwata H, Szele FG, Isoda H. Rosemary extract activates oligodendrogenesis genes in mouse brain and improves learning and memory ability. Biomed Pharmacother 2024; 179:117350. [PMID: 39197189 DOI: 10.1016/j.biopha.2024.117350] [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: 06/17/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024] Open
Abstract
Rosemary (Rosmarinus officinalis L.) is a rich source of dietary bioactive compounds such as rosmarinic acid and carnosol with a large repertoire of pharmacological properties, including anti-inflammatory and neuroprotective activities. In the present study, we investigated rosemary as a potential new therapeutic agent for cognitive function and other symptoms of aging. In this present study, we have aimed to investigate the effects of oral administration of rosemary extract (RME) on learning and memory in the context of other biomarkers-related cognitive function and neurotransmitter levels in senescent accelerated prone 8 (SAMP8) mouse, a model of accelerating aging and Alzheimer's disease. The Morris water maze (MWM) test showed improved spatial learning and memory behavior in RME treated SAMP8 mouse. Moreover, RME decreased Aβ42 and inflammatory cytokine levels and increased BDNF, Sirt1, and neurotransmitter levels in SAMP8 mouse. Whole-genome microarray analysis revealed that RME significantly increased gene expression related to oligodendrocyte differentiation, myelination, and ATP production in the hippocampus and decreased gene expression related to stress, neuroinflammation, and apoptosis. Also, in the SAMP8 hippocampus, RME significantly increased Olig1 and Olig2 expression. Altogether, our study is the first to report improvement of spatial learning and memory of RME, modulation of genes important for oligodendrogenesis, and Anti-neuroinflammatory effect by suppressing Aβ42 levels in mouse brain and thus highlights the prospects of RME in the treatment of cognitive dysfunction and aging.
Collapse
Affiliation(s)
- Kazunori Sasaki
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan; AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, Tsukuba, Japan
| | - Jemima Becker
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Jun Ong
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Sabina Ciaghi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Lynn S Guldin
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Sofia Savastano
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Satoshi Fukumitsu
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tsukuba, Japan
| | - Hidetoshi Kuwata
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan; AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, Tsukuba, Japan; Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tsukuba, Japan; Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
124
|
Zhvania MG, Japaridze N, Tizabi Y, Pochkhidze N, Lobzhanidze G. Effects of high-intensity chronic noise on spatial memory in male versus female rats. Eur J Neurosci 2024; 60:5581-5590. [PMID: 39180282 DOI: 10.1111/ejn.16514] [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: 03/21/2024] [Revised: 07/25/2024] [Accepted: 08/10/2024] [Indexed: 08/26/2024]
Abstract
The detrimental effects of high-intensity noise on the auditory system and emotional status, including the induction of anxiety, are well documented. Preclinical as well as epidemiological and clinical studies have solidly established differential responses between males and females to various stressful stimuli, including high-intensity white noise (HIWN). However, whether chronic exposure to noise affects cognitive functions and whether this effect is sex dependent has not been adequately addressed. In this study, we used two cognitive test paradigms, such as the Morris water maze (MWM) and the multi-branch maze (MBM), to test the effect of chronic HIWN on indices of spatial learning and memory in both male and female Wistar rats. Our findings indicate that daily (1 h) exposure to 100 dB of noise for 30 consecutive days induces different task-dependent responses in male versus female rats. For example, in the acquisition phase of MWM, female rats exposed to noise outperformed their male counterparts at twice the speed. Similarly, in the MBM test, noise-exposed female rats outperformed the male rats in reaching the nest box. It is clear from these studies that noise impairs cognitive functions twice as negatively in male rats as in female rats. Thus, sex-related differences in spatial learning and memory in response to HIWN must be taken into consideration when investigating the neurobiological components and/or treatment modalities.
Collapse
Affiliation(s)
- Mzia G Zhvania
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Nadezhda Japaridze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
- Carl Zeiss Scientific and Education Center, New Vision University, Tbilisi, Georgia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Nino Pochkhidze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Giorgi Lobzhanidze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| |
Collapse
|
125
|
Sun H, Cai R, Li R, Li M, Gao L, Li X. Conjunctive processing of spatial border and locomotion in retrosplenial cortex during spatial navigation. J Physiol 2024; 602:5017-5038. [PMID: 39216077 DOI: 10.1113/jp286434] [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: 03/04/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Spatial information and dynamic locomotor behaviours are equally important for achieving locomotor goals during spatial navigation. However, it remains unclear how spatial and locomotor information is integrated during the processing of self-initiated spatial navigation. Anatomically, the retrosplenial cortex (RSC) has reciprocal connections with brain regions related to spatial processing, including the hippocampus and para-hippocampus, and also receives inputs from the secondary motor cortex. In addition, RSC is functionally associated with allocentric and egocentric spatial targets and head-turning. So, RSC may be a critical region for integrating spatial and locomotor information. In this study, we first examined the role of RSC in spatial navigation using the Morris water maze and found that mice with inactivated RSC took a longer time and distance to reach their destination. Then, by imaging neuronal activity in freely behaving mice within two open fields of different sizes, we identified a large proportion of border cells, head-turning cells and locomotor speed cells in the superficial layer of RSC. Interestingly, some RSC neurons exhibited conjunctive coding for both spatial and locomotor signals. Furthermore, these conjunctive neurons showed higher prediction accuracy compared with simple spatial or locomotor neurons in special navigator scenes using the border, turning and positive-speed conjunctive cells. Our study reveals that the RSC is an important conjunctive brain region that processes spatial and locomotor information during spatial navigation. KEY POINTS: Retrosplenial cortex (RSC) is indispensable during spatial navigation, which was displayed by the longer time and distance of mice to reach their destination after the inactivation of RSC in a water maze. The superficial layer of RSC has a larger population of spatial-related border cells, and locomotion-related head orientation and speed cells; however, it has few place cells in two-dimensional spatial arenas. Some RSC neurons exhibited conjunctive coding for both spatial and locomotor signals, and the conjunctive neurons showed higher prediction accuracy compared with simple spatial or locomotor neurons in special navigation scenes. Our study reveals that the RSC is an important conjunctive brain region that processes both spatial and locomotor information during spatial navigation.
Collapse
Affiliation(s)
- Hao Sun
- Department of Neurology of the Second Affiliated Hospital and Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China
- Nanhu Brain-computer Interface Institute, Hangzhou, China
- Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Ruolan Cai
- Department of Neurology of the Second Affiliated Hospital and Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China
| | - Rui Li
- Department of Neurology of the Second Affiliated Hospital and Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Lixia Gao
- Department of Neurology of the Second Affiliated Hospital and Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China
- Nanhu Brain-computer Interface Institute, Hangzhou, China
- Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Xinjian Li
- Department of Neurology of the Second Affiliated Hospital and Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Neurobiology of Zhejiang Province, Hangzhou, China
| |
Collapse
|
126
|
Li X, Long J, Yao C, Liu X, Li N, Zhou Y, Li D, Xiong G, Wang K, Hao Y, Chen K, Zhou Z, Ji A, Luo P, Cai T. The role of BTG2/PI3K/AKT pathway-mediated microglial activation in T-2 toxin-induced neurotoxicity. Toxicol Lett 2024; 400:81-92. [PMID: 39147216 DOI: 10.1016/j.toxlet.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 07/07/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
T-2 toxin is one of the mycotoxins widely distributed in human food and animal feed. Our recent work has shown that microglial activation may contribute to T-2 toxin-induced neurotoxicity. However, the molecular mechanisms involved need to be further clarified. To address this, we employed high-throughput transcriptome sequencing and found altered B cell translocation gene 2 (BTG2) expression levels in microglia following T-2 toxin treatment. It has been shown that altered BTG2 expression is involved in a range of neurological pathologies, but whether it's involved in the regulation of microglial activation is unclear. The aim of this study was to investigate the role of BTG2 in T-2 toxin-induced microglial activation. The results of animal experiments showed that T-2 toxin caused neurobehavioral disorders and promoted the expression of microglial BTG2 and pro-inflammatory activation of microglia in hippocampus and cortical, while microglial inhibitor minocycline inhibited these changes. The results of in vitro experiments showed that T-2 toxin enhanced BTG2 expression and pro-inflammatory microglial activation, and inhibited BTG2 expression weakened T-2 toxin-induced microglial activation. Moreover, T-2 toxin activated PI3K/AKT and its downstream NF-κB signaling pathway, which could be reversed after knock-down of BTG2 expression. Meanwhile, the PI3K inhibitor LY294002 also blocked this process. Therefore, BTG2 may be involved in T-2 toxin's ability to cause microglial activation through PI3K/AKT/NF-κB pathway.
Collapse
Affiliation(s)
- Xiukuan Li
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jinyun Long
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chunyan Yao
- Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiaoling Liu
- Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Na Li
- Chongqing Yongchuan District Center for Disease Control and Prevention, Chongqing 402160, China
| | - Yumeng Zhou
- Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Dawei Li
- Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Guiyuan Xiong
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Kexue Wang
- Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuhui Hao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ka Chen
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ziyuan Zhou
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ailing Ji
- Department of Preventive Medicine, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.
| | - Peng Luo
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China.
| | - Tongjian Cai
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
127
|
Guan Y, Wang C, Li L, Dai X, Liu Y, Hsiang T, Liu S, Wang D. Structural characterization of Hericium coralloides polysaccharide and its neuroprotective function in Alzheimer's disease. Int J Biol Macromol 2024; 277:133865. [PMID: 39019356 DOI: 10.1016/j.ijbiomac.2024.133865] [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: 04/03/2024] [Revised: 06/21/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder. Polysaccharides have been scientifically demonstrated to possess neuroprotective properties. In this study, a polysaccharide was isolated from the fruiting bodies of Hericium coralloides using hot water extraction and purified using column chromatography. This H. coralloides polysaccharide (HCP) is a galactan with a main chain of →6)-α-d-Galp-(1 → and a molecular weight of 16.06 kDa. The partial α-l-Fucp-(1 → substitution takes place at its O-2 position. The neuroprotective effects of HCP were investigated in an APP/PS1 mouse model of Alzheimer's disease. The step-down and Morris water maze tests demonstrated that HCP effectively ameliorated cognitive impairment. After 8-week treatment, HCP reduced amyloid-β plaques and phosphorylated tau protein deposition. In combination with the gut microbiota and metabolites, proteomic analysis suggested that the neuroprotective effects of HCP are associated with neuroinflammation and autophagy. Immunofluorescence and western blotting analyses confirmed that HCP facilitated the polarization of M2 microglia by augmenting autophagy flux, thereby effectively reducing levels of amyloid-β plaques and neuroinflammation. These data demonstrate that HCP effectively mitigates neuroinflammation by enhancing autophagic flux, demonstrating its potential for the treatment of AD.
Collapse
Affiliation(s)
- Yue Guan
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| | - Chunyue Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| | - Xiaojing Dai
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| | - Yang Liu
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| | - Tom Hsiang
- School of Environmental Sciences, University of Guelph, Ontario N1G 2W1, Guelph, Canada.
| | - Shuyan Liu
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| | - Di Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China; School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
128
|
Chen R, Pan C, Mao X, Zhang Y, Chen G, Xu M, Nivar J, Tao Y, Cao H, Li J. Chloride intracellular channel 4 blockade improves cognition in mice with Alzheimer's disease: CLIC4 protein expression and tau protein hyperphosphorylation. Int J Biol Macromol 2024; 278:134972. [PMID: 39181373 DOI: 10.1016/j.ijbiomac.2024.134972] [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: 06/28/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Numerous academic literature suggests that amyloid-β (Aβ) deposition, tau protein phosphorylation, and irreversible neuronal death are the three major causes of AD. The chloride intracellular channel (CLIC) protein family not only regulates the polarisation of neurons, but also has important implications for neuronal survival. Chloride intracellular channel 4 (CLIC4) can be pathologically activated by cyclin-dependent kinase 5 (Cdk5), which causes a significant increase in the expression of CLIC4 and mediates neuronal apoptosis. CLIC4 knockdown inhibits H2O2-induced neuronal apoptosis; however, the relationship between CLIC4 and AD remains unknown. In the present study, we showed that CLIC4 expression was elevated in the hippocampus of AD mice; knockdown of hippocampal CLIC4 alleviated Aβ25-35-induced cognitive impairment in mice; overexpression of hippocampal CLIC4 accelerated Aβ deposition and tau protein hyperphosphorylation in young AD mice (APP/PS1 mice at three months of age). CLIC4 overexpressing mice had a longer escape latency compared to controls in behavioural testing (Morris water maze and T-maze tests). By Co-immunoprecipitation/mass spectrometry (Co-IP/MS) of HT22 cells to identify proteins that specifically bind to CLIC4, we found interactions with CCAAT enhancer binding protein (C/EBPβ); a critical pathway involved in the development of various neurodegenerative diseases. In addition, the knockdown of hippocampal CLIC4 alleviated AD-like pathology by inhibiting the C/EBPβ/AEP signaling pathway. These data suggest an essential role for high CLIC4 expression in the pathophysiology of AD and reveal that inhibition of CLIC4 expression may provide an opportunity for treatment.
Collapse
Affiliation(s)
- Rui Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China; The Second Affiliated Hospital Zhejiang University School of Medicine, Department of Anesthesiology, Hangzhou 310000, Zhejiang Province, China
| | - Chi Pan
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - Xinyu Mao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - Yantong Zhang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - Gang Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - Mengting Xu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - John Nivar
- Rutgers New Jersey Medical School, Department of Anesthesiology, Newark, NJ, USA
| | - Yuanxiang Tao
- Rutgers New Jersey Medical School, Department of Anesthesiology, Newark, NJ, USA
| | - Hong Cao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China
| | - Jun Li
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Department of Anesthesiology, Wenzhou 325000, Zhejiang Province, China.
| |
Collapse
|
129
|
Abedi A, Foroutan T, Mohaghegh Shalmani L, Dargahi L. Sex-dependent susceptibility to brain metabolic dysfunction and memory impairment in response to pre and postnatal high-fat diet. J Nutr Biochem 2024; 132:109675. [PMID: 38945454 DOI: 10.1016/j.jnutbio.2024.109675] [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: 02/15/2024] [Revised: 05/15/2024] [Accepted: 05/31/2024] [Indexed: 07/02/2024]
Abstract
The developing brain is sensitive to the impacts of early-life nutritional intake. This study investigates whether maternal high fat diet (HFD) causes glucose metabolism impairment, neuroinflammation, and memory impairment in immature and adult offspring, and whether it may be affected by postweaning diets in a sex-dependent manner in adult offspring. After weaning, female rats were fed HFD (55.9% fat) or normal chow diet (NCD; 10% fat) for 8 weeks before mating, during pregnancy, and lactation. On postnatal day 21 (PND21), the male and female offspring of both groups were split into two new groups, and NCD or HFD feeding was maintained until PND180. On PND21 and PND180, brain glucose metabolism, inflammation, and Alzheimer's pathology-related markers were by qPCR. In adult offspring, peripheral insulin resistance parameters, spatial memory performance, and brain glucose metabolism (18F-FDG-PET scan and protein levels of IDE and GLUT3) were assessed. Histological analysis was also performed on PND21 and adult offspring. On PND21, we found that maternal HFD affected transcript levels of glucose metabolism markers in both sexes. In adult offspring, more profoundly in males, postweaning HFD in combination with maternal HFD induced peripheral and brain metabolic disturbances, impaired memory performance and elevated inflammation, dementia risk markers, and neuronal loss. Our results suggest that maternal HFD affects brain glucose metabolism in the early ages of both sexes. Postweaning HFD sex-dependently causes brain metabolic dysfunction and memory impairment in later-life offspring; effects that can be worsened in combination with maternal HFD.
Collapse
Affiliation(s)
- Azam Abedi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Mohaghegh Shalmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
130
|
Aziz N, Wal P, Patel A, Prajapati H. A comprehensive review on the pharmacological role of gut microbiome in neurodegenerative disorders: potential therapeutic targets. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7307-7336. [PMID: 38734839 DOI: 10.1007/s00210-024-03109-4] [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: 10/09/2023] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Neurological disorders, including Alzheimer and Parkinson's, pose significant challenges to public health due to their complex etiologies and limited treatment options. Recent advances in research have highlighted the intricate bidirectional communication between the gut microbiome and the central nervous system (CNS), revealing a potential therapeutic avenue for neurological disorders. Thus, this review aims to summarize the current understanding of the pharmacological role of gut microbiome in neurological disorders. Mounting evidence suggests that the gut microbiome plays a crucial role in modulating CNS function through various mechanisms, including the production of neurotransmitters, neuroactive metabolites, and immune system modulation. Dysbiosis, characterized by alterations in gut microbial composition and function, has been observed in many neurological disorders, indicating a potential causative or contributory role. Pharmacological interventions targeting the gut microbiome have emerged as promising therapeutic strategies for neurological disorders. Probiotics, prebiotics, antibiotics, and microbial metabolite-based interventions have shown beneficial effects in animal models and some human studies. These interventions aim to restore microbial homeostasis, enhance microbial diversity, and promote the production of beneficial metabolites. However, several challenges remain, including the need for standardized protocols, identification of specific microbial signatures associated with different neurological disorders, and understanding the precise mechanisms underlying gut-brain communication. Further research is necessary to unravel the intricate interactions between the gut microbiome and the CNS and to develop targeted pharmacological interventions for neurological disorders.
Collapse
Affiliation(s)
- Namra Aziz
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India
| | - Pranay Wal
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India.
| | - Aman Patel
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India
| | - Harshit Prajapati
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India
| |
Collapse
|
131
|
Jha AB, Chaube UJ, Jha AB. Ellagic acid improves the symptoms of early-onset Alzheimer's disease: Behavioral and physiological correlates. Heliyon 2024; 10:e37372. [PMID: 39309887 PMCID: PMC11416286 DOI: 10.1016/j.heliyon.2024.e37372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/27/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Oryza sativa is a globally recognized staple food, rich in essential phyto-phenolic compounds such as γ-Oryzanol (OZ), Ferulic acid (FA), and Ellagic acid (EA). These phytochemicals are known for their potential to beneficially modulate molecular biochemistry. The present investigation aimed to evaluate the neuroprotective and cognitive enhancement effects of Oryza sativa phyto-phenolics in a model of early-onset Alzheimer's disease (EOAD) induced by Aβ (1-42) in animals. In-silico studies suggested that FA, OZ, and EA have target specificity for Aβ, with EA being further selected based on its potent in-vitro Aβ anti-aggregatory effects for exploring neurodegenerative conditions. The in-vivo experiments demonstrated that EA exerts therapeutic effects in Aβ-induced EOAD, modulating both biochemical and behavioral outcomes. EA treatment at two dose levels, EA70 and EA140 (70 μM and 140 μM, respectively, administered i.c.v.), significantly counteracted Aβ aggregation and modulated the Ca2⁺/Calpain/GSK-3β/CDK5 signaling pathways, exhibiting anti-tauopathy effects. Additionally, EA was shown to exert anti-inflammatory effects by preventing astroglial activation, modulating FAIM-L expression, and protecting against TNF-α-induced apoptotic signals. Moreover, the neuromodulatory effects of EA were attributed to the regulation of CREB levels, Dnm-1 expression, and synaptophysin levels, thereby enhancing LTP and synaptic plasticity. EA also induced beneficial cytological and behavioral changes, improving both long-term and short-term spatial memory as well as associative learning behavior in the animal model, which underscores its cognitive enhancement properties.
Collapse
Affiliation(s)
- Abhishek B. Jha
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Udit J. Chaube
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | | |
Collapse
|
132
|
Ali M, Din Wani SU, Manjula S, Mruthunjaya K, Shakeel F, DR B, Sridhar SB, Mohiuddin I, Mir RH, Dey T. Divine noni's protective impact on Swiss albino mice's short-term memory impairment caused by cyclophosphamide: A behavioral and biochemical approach. Heliyon 2024; 10:e37557. [PMID: 39309823 PMCID: PMC11415706 DOI: 10.1016/j.heliyon.2024.e37557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/26/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
Cyclophosphamide (CYL) is a first-line cancer chemotherapeutic agent widely used for the treatment of cancer that has severe toxic effects. The primary mechanism by which CYL induces toxicity through free radical generation. Morinda citrifolia (Noni) fruit juice is an herbal remedy documented to have antioxidant properties. The aim of the current study was to investigate the protective effect of noni against CYL-induced memory impairment in Swiss albino mice. Treatment schedule: Group 1: Normal: Received vehicle; Group 2: CYL treatment: Received CYL (40.0 mg/kg b.w. i.p.) on day one; Group 3: NJ treatment: Received NJ (360 mg/b.w. p.o.) once daily for 14 days. Group 4: DNG treatment: DNG (360 mg/b.w. p.o.) once daily for 14 days, Group 5: NJ + CYL treatment: Received CYL (40.0 mg/kg b.w. i.p.) on day one and after half an hour of received NJ (360 mg/b.w. p.o.) once daily for 14 days. Group 6: DNG + CYL treatment: Received CYL (40.0 mg/kg b.w. i.p.) on day one and after half an hour received DNG (360 mg/b.w. p.o.) once daily for 14 days. Mice were subjected to the Morris water maze (MWM) challenge for two weeks as part of a behavioral study. Short-term memory impairment was observed in the behavioral activity of CYL-treated mice in the MWM test in the 1st week trial, and this effect was reversed in the 2nd week trial in the combination treatment group. The behavioral analysis proved that noni supplementation reduced the risk of memory impairment caused by CYL. Biochemical analysis revealed that CYL markedly increased the levels of AChE and MDA in brain tissue. Similarly, decreases in the levels of antioxidants, i.e., GSH, CAT, SOD and GST, were detected in the brain tissue of the mice exposed to CYL. Qualitative and quantitative examinations of histopathological examination of the mouse hippocampus supported the above findings. The results demonstrated that noni supplement therapy reversed the changes in the MDA, AChE, and antioxidant enzyme levels while improving the behavioral and histological alterations caused by CYL. Long-term hippocampal growth and memory are unaffected, suggesting that CYL is less harmful. According to our research, supplementing with noni in conjunction with CYL may be a helpful treatment strategy for treating memory impairment caused by CYL.
Collapse
Affiliation(s)
- Mohammad Ali
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
- Department of Pharmacology, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G Nagar, Bellur, Karnataka, 571418, India
| | - Shahid Ud Din Wani
- Department of Pharmaceutical Sciences, School of Applied Science and Technology, University of Kashmir, Srinagar, 190006, India
| | - S.N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - K. Mruthunjaya
- Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher, Education and Research, Mysuru, 570015, India
| | - Faiyaz Shakeel
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Bharathi DR
- Department of Pharmacology, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G Nagar, Bellur, Karnataka, 571418, India
| | - Sathvik B. Sridhar
- Department of Clinical Pharmacy and Pharmacology, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, PO Box, 11172, United Arab Emirates
| | - Ishfaq Mohiuddin
- Department of Zoology, Annamalai University, Annamalainagar, 608002, India
| | - Reyaz Hassan Mir
- Department of Pharmaceutical Sciences, School of Applied Science and Technology, University of Kashmir, Srinagar, 190006, India
| | - Tathagata Dey
- Department of Pharmaceutical Chemistry, East Point College of Pharmacy, Bangalore, 560049, India
| |
Collapse
|
133
|
Katimbwa DA, Kim Y, Kim MJ, Jeong M, Lim J. Solubilized β-Glucan Supplementation in C57BL/6J Mice Dams Augments Neurodevelopment and Cognition in the Offspring Driven by Gut Microbiome Remodeling. Foods 2024; 13:3102. [PMID: 39410136 PMCID: PMC11476385 DOI: 10.3390/foods13193102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
A maternal diet rich in dietary fiber, such as β-glucan, plays a crucial role in the offspring's acquisition of gut microbiota and the subsequent shaping of its microbiome profile and metabolome. This in turn has been shown to aid in neurodevelopmental processes, including early microglial maturation and immunomodulation via metabolites like short chain fatty acids (SCFAs). This study aimed to investigate the effects of oat β-glucan supplementation, solubilized by citric acid hydrolysis, from gestation to adulthood. Female C57BL/6J mice were orally supplemented with soluble oat β-glucan (ObG) or carboxymethyl cellulose (CMC) via drinking water at 200 mg/kg body weight during breeding while the control group received 50 mg/kg body weight of carboxymethyl cellulose. ObG supplementation increased butyrate production in the guts of both dams and 4-week-old pups, attributing to alterations in the gut microbiota profile. One-week-old pups from the ObG group showed increased neurodevelopmental markers similar to four-week-old pups that also exhibited alterations in serum markers of metabolism and anti-inflammatory cytokines. Notably, at 8 weeks, ObG-supplemented pups exhibited the highest levels of spatial memory and cognition compared to the control and CMC groups. These findings suggest a potential enhancement of neonatal neurodevelopment via shaping of early-life gut microbiome profile, and the subsequent increased later-life cognitive function.
Collapse
Affiliation(s)
- Dorsilla A. Katimbwa
- Department of Food Biomaterials, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Yoonsu Kim
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Min Jeong Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Minsoo Jeong
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jinkyu Lim
- Department of Food Biomaterials, Kyungpook National University, Daegu 41566, Republic of Korea;
| |
Collapse
|
134
|
Lin L, Xu M, Ma J, Du C, Zang Y, Huang A, Wei C, Gao Q, Gan S. Behavioral Assessment Reveals GnRH Immunocastration as a Better Alternative to Surgical Castration. Animals (Basel) 2024; 14:2796. [PMID: 39409745 PMCID: PMC11475323 DOI: 10.3390/ani14192796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Castration is often employed in animal management for reproductive control. However, it is important to evaluate its impact on animal welfare. In this study, we developed rat models for both surgical (n = 6) and GnRH immunocastration (n = 6) to assess the effects of these castration methods on physiological and behavioral characteristics. The novel GnRH-based vaccine significantly increased serum GnRH antibody levels and drastically reduced testosterone, with the testes shrinking to one-fifth the size of those in the control group, thereby halting spermatogenesis at the secondary spermatocyte stage. Behavioral evaluations demonstrated that sexual behavior was significantly suppressed in both surgically and immunologically castrated groups compared to the control, confirming the effectiveness of both methods. However, psychological tests revealed significant signs of depression and social deficits in the surgically castrated group, whereas the behavior of the GnRH-immunocastrated group did not significantly differ from the control. Furthermore, no significant differences in learning and memory were observed among the three groups in the water maze test. Compared to surgical castration, GnRH immunocastration offers effective results and better animal welfare, providing a more humane alternative for livestock management.
Collapse
Affiliation(s)
- Liuxia Lin
- College of Life Science, Tarim University, Alaer 843300, China; (L.L.); (A.H.)
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.M.); (C.D.); (C.W.)
| | - Mengsi Xu
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Sciences, Shihezi 832000, China;
| | - Jian Ma
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.M.); (C.D.); (C.W.)
| | - Chunmei Du
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.M.); (C.D.); (C.W.)
| | - Yaxin Zang
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China;
| | - Amei Huang
- College of Life Science, Tarim University, Alaer 843300, China; (L.L.); (A.H.)
| | - Chen Wei
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.M.); (C.D.); (C.W.)
| | - Qinghua Gao
- College of Animal Science and Technology, Tarim University, Alaer 843300, China
| | - Shangquan Gan
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.M.); (C.D.); (C.W.)
| |
Collapse
|
135
|
Nakhal MM, Yassin LK, Alyaqoubi R, Saeed S, Alderei A, Alhammadi A, Alshehhi M, Almehairbi A, Al Houqani S, BaniYas S, Qanadilo H, Ali BR, Shehab S, Statsenko Y, Meribout S, Sadek B, Akour A, Hamad MIK. The Microbiota-Gut-Brain Axis and Neurological Disorders: A Comprehensive Review. Life (Basel) 2024; 14:1234. [PMID: 39459534 PMCID: PMC11508655 DOI: 10.3390/life14101234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Microbes have inhabited the earth for hundreds of millions of years longer than humans. The microbiota-gut-brain axis (MGBA) represents a bidirectional communication pathway. These communications occur between the central nervous system (CNS), the enteric nervous system (ENS), and the emotional and cognitive centres of the brain. The field of research on the gut-brain axis has grown significantly during the past two decades. Signalling occurs between the gut microbiota and the brain through the neural, endocrine, immune, and humoral pathways. A substantial body of evidence indicates that the MGBA plays a pivotal role in various neurological diseases. These include Alzheimer's disease (AD), autism spectrum disorder (ASD), Rett syndrome, attention deficit hyperactivity disorder (ADHD), non-Alzheimer's neurodegeneration and dementias, fronto-temporal lobe dementia (FTLD), Wilson-Konovalov disease (WD), multisystem atrophy (MSA), Huntington's chorea (HC), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), temporal lobe epilepsy (TLE), depression, and schizophrenia (SCZ). Furthermore, the bidirectional correlation between therapeutics and the gut-brain axis will be discussed. Conversely, the mood of delivery, exercise, psychotropic agents, stress, and neurologic drugs can influence the MGBA. By understanding the MGBA, it may be possible to facilitate research into microbial-based interventions and therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Mohammed M. Nakhal
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Lidya K. Yassin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Rana Alyaqoubi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Sara Saeed
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Alreem Alderei
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Alya Alhammadi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Mirah Alshehhi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Afra Almehairbi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Shaikha Al Houqani
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Shamsa BaniYas
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Haia Qanadilo
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Safa Shehab
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| | - Yauhen Statsenko
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
- Neuroscience Platform, ASPIRE Precision Medicine Institute in Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Sarah Meribout
- Internal Medicine Department, Maimonides Medical Center, New York, NY 11219, USA;
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Bo Box 15551, United Arab Emirates; (B.S.); (A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 1551, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Bo Box 15551, United Arab Emirates; (B.S.); (A.A.)
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates (S.B.); (S.S.)
| |
Collapse
|
136
|
Li Y, Tang C, Song Y. Protocol to establish a demyelinated animal model to study hippocampal neurogenesis and cognitive function in adult rodents. STAR Protoc 2024; 5:103242. [PMID: 39093706 PMCID: PMC11342265 DOI: 10.1016/j.xpro.2024.103242] [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: 06/02/2024] [Revised: 06/30/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024] Open
Abstract
Cognitive dysfunction is a prevalent feature in multiple sclerosis, a chronic inflammatory demyelinating disease, which may be correlated with the impairment of adult hippocampal neurogenesis. Here, we present a detailed protocol for the induction of cuprizone demyelinated mice to assess the cognitive function and explore the precise mechanisms underlying cognitive deficits in demyelinated hippocampus. We describe steps for behavioral tests, 5-Ethynyl-2'-deoxyuridine (EdU) and bromodeoxyuridine (BrdU) administration, retrovirus packaging and stereotactic injection, hippocampal tissue preparation, and immunofluorescence staining. For complete details on the use and execution of this protocol, please refer to Song et al.1.
Collapse
Affiliation(s)
- Yuhan Li
- Department of Neurology, The Third Affiliated Hospital of SUN Yat-sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China
| | - Changyong Tang
- Department of Neurology, The Third Affiliated Hospital of SUN Yat-sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China.
| | - Yanna Song
- Department of Neurology, The Third Affiliated Hospital of SUN Yat-sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong Province, China.
| |
Collapse
|
137
|
Parrini M, Caroni P, Spolidoro M. Protocol to investigate the gradual selection and deployment of goal-oriented search strategies during unsupervised navigation in mice. STAR Protoc 2024; 5:103290. [PMID: 39226172 PMCID: PMC11419921 DOI: 10.1016/j.xpro.2024.103290] [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: 04/15/2024] [Revised: 06/29/2024] [Accepted: 08/14/2024] [Indexed: 09/05/2024] Open
Abstract
The ability of rodents to effectively navigate in an environment is based on trial-and-error learning and flexible decision-making and can be analyzed via navigational trajectories. We present a protocol for studying the deployment of search strategies in mice using the Morris water maze. We describe steps for assigning mice to different maze variations and procedures for post-training tracking and analysis. This protocol represents an effective behavioral readout to probe brain networks involved in strategy deployment and goal-oriented behavior. For complete details on the use and execution of this protocol, please refer to Parrini et al.1.
Collapse
Affiliation(s)
- Martina Parrini
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Pico Caroni
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.
| | - Maria Spolidoro
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.
| |
Collapse
|
138
|
Fang Y, Shen P, Xu L, Shi Y, Wang L, Yang M. PDTC improves cognitive impairment in LPS-induced ARDS by regulating miR-181c/NF-κB axis-mediated neuroinflammation. Brain Inj 2024; 38:918-927. [PMID: 38828532 DOI: 10.1080/02699052.2024.2361623] [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: 08/30/2022] [Revised: 04/02/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Cognitive impairment is a severe complication of acute respiratory distress syndrome (ARDS). Emerging studies have revealed the effects of pyrrolidine dithiocarbamate (PDTC) on improving surgery-induced cognitive impairment. The major aim of the study was to investigate whether PDTC protected against ARDS-induced cognitive dysfunction and to identify the underlying mechanisms involved. METHODS The rat model of ARDS was established by intratracheal instillation of lipopolysaccharide (LPS), followed by treatment with PDTC. The cognitive function of rats was analyzed by the Morris Water Maze, and pro-inflammatory cytokines were assessed by quantitative real-time PCR, enzyme-linked immunosorbent assay, and western blot assays. A dual-luciferase reporter gene assay was performed to identify the relationship between miR-181c and its target gene, TAK1 binding protein 2 (TAB2). RESULTS The results showed that PDTC improved cognitive impairment and alleviated neuroinflammation in the hippocampus in LPS-induced ARDS model. Furthermore, we demonstrated that miR-181c expression was downregulated in the hippocampus of the ARDS rats, which was restored by PDTC treatment. In vitro studies showed that miR-181c alleviated LPS-induced pro-inflammatory response by inhibiting TAB2, a critical molecule in the nuclear factor (NF)-κB signaling pathway. CONCLUSION PDTC improves cognitive impairment in LPS-induced ARDS by regulating miR-181c/NF-κB axis-mediated neuroinflammation, providing a potential opportunity for the treatment of this disease.
Collapse
Affiliation(s)
- Ying Fang
- Department of Pathology, The First Hospital of Jiaxing & Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Peng Shen
- Department of Intensive Care Unit, The First Hospital of Jiaxing & Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Longsheng Xu
- Department of Central Laboratory, The First Hospital of Jiaxing & Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yunchao Shi
- Department of Intensive Care Unit, The First Hospital of Jiaxing & Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Liyan Wang
- Department of General Practice, The First Hospital of Jiaxing & Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Maoxian Yang
- Department of Intensive Care Unit, The First Hospital of Jiaxing & Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
139
|
Li S, Liu H, Lv P, Yao Y, Peng L, Xia T, Yan C, Ma Z, Chen ZP, Zhao C, Gu X. Microglia mediate memory dysfunction via excitatory synaptic elimination in a fracture surgery mouse model. J Neuroinflammation 2024; 21:227. [PMID: 39285282 PMCID: PMC11406843 DOI: 10.1186/s12974-024-03216-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024] Open
Abstract
Cognitive impairment is a common issue among human patients undergoing surgery, yet the neural mechanism causing this impairment remains unidentified. Surgical procedures often lead to glial cell activation and neuronal hypoexcitability, both of which are known to contribute to postoperative cognitive dysfunction (POCD). However, the role of neuron-glia crosstalk in the pathology of POCD is still unclear. Through integrated transcriptomics and proteomics analyses, we found that the complement cascades and microglial phagocytotic signaling pathways are activated in a mouse model of POCD. Following surgery, there is a significant increase in the presence of complement C3, but not C1q, in conjunction with presynaptic elements. This triggers a reduction in excitatory synapses, a decline in excitatory synaptic transmission, and subsequent memory deficits in the mouse model. By genetically knockout out C3ar1 or inhibiting p-STAT3 signaling, we successfully prevented neuronal hypoexcitability and alleviated cognitive impairment in the mouse model. Therefore, targeting the C3aR and downstream p-STAT3 signaling pathways could serve as potential therapeutic approaches for mitigating POCD.
Collapse
Affiliation(s)
- Shuming Li
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Huan Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Pin Lv
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yu Yao
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Liangyu Peng
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tianjiao Xia
- Medical School, Nanjing University, Nanjing, China
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhang-Peng Chen
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
140
|
Jan A, Shah M, Shah SA, Habib SH, Ehtesham E, Ahmed N. Melatonin rescues pregnant female mice and their juvenile offspring from high fat diet-induced alzheimer disease neuropathy. Heliyon 2024; 10:e36921. [PMID: 39281480 PMCID: PMC11395765 DOI: 10.1016/j.heliyon.2024.e36921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
High fat diet (HFD) is a prime factor, which contributes to the present epidemic of metabolic syndrome. Prolonged intake of HFD induces oxidative stress (OS) that in turn causes neuroinflammation, neurodegeneration, insulin resistance, amyloid burden, synaptic dysfunction and cognitive impairment hence leading to Alzheimer's disease neuropathy. Melatonin (secreted by the Pineal gland) has the potential to nullify the toxic effects of reactive oxygen species (ROS) and have been shown to ameliorate various complications induced by HFD in rodent models. This study aimed to assess the neurotherapeutic effects of melatonin on HFD-induced neuroinflammation and neurodegeneration mediated by OS in pregnant female mice and their offspring. Western blotting, immunohistochemistry and antioxidant enzyme assays were used for quantification of samples from the hippocampal region of the brain of pregnant albino mice and their offspring. Short- and long-term memory was assessed by Y-maze and Morris Water Maze tests. HFD significantly induced OS leading to AD like neuropathology in the pregnant mice and their offspring while melatonin administration simultaneously with the HFD significantly prevented this neuropathy. This study reports that melatonin exerts these effects through the stimulation of SIRT1/Nrf2/HO-1 pathway that in turn reduces the HFD-induced OS and its downstream signaling. In conclusion melatonin prevents HFD-induced multiple complications that ultimately leads to the memory dysfunction in pregnant female mice and their successive generation via activation of SIRT1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Amin Jan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Mohsin Shah
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Shahid Ali Shah
- Department of Biochemistry, Haripur University, Haripur, Pakistan
| | - Syed Hamid Habib
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Ehtesham Ehtesham
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Naseer Ahmed
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| |
Collapse
|
141
|
Dai WB, Zhang X, Jiang XL, Zhang YZ, Chen LK, Tian WT, Zhou XX, Sun XY, Huang LL, Gu XY, Chen XM, Wu XD, Tian J, Yu WF, Shen L, Su DS. The kynurenine pathway regulated by intestinal innate lymphoid cells mediates postoperative cognitive dysfunction. Mucosal Immunol 2024:S1933-0219(24)00095-3. [PMID: 39251184 DOI: 10.1016/j.mucimm.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Postoperative cognitive dysfunction (POCD) is a prevalent neurological complication that can impair learning and memory for days, months, or even years after anesthesia/surgery. POCD is strongly associated with an altered composition of the gut microbiota (dysbiosis), but the accompanying metabolic changes and their role in gut-brain communication and POCD pathogenesis remain unclear. Here, the present study reports that anesthesia/surgery in aged mice induces elevated intestinal indoleamine 2,3-dioxygenase (IDO) expression and activity, which shifts intestinal tryptophan (TRP) metabolism toward more IDO-catalyzed kynurenine (KYN) and less gut bacteria-catabolized indoleacetic acid (IAA). Both anesthesia/surgery and intraperitoneal KYN administration induce increased KYN levels that correlate with impaired spatial learning and memory, whereas dietary IAA supplementation attenuates the anesthesia/surgery-induced cognitive impairment. Mechanistically, anesthesia/surgery increases interferon-γ (IFN-γ)-producing group 1 innate lymphoid cells (ILC1) in the small intestine lamina propria and elevates intestinal IDO expression and activity, as indicated by the higher ratio of KYN to TRP. The IDO inhibitor 1-MT and antibodies targeting IFN-γ or ILCs mitigate anesthesia/surgery-induced cognitive dysfunction, suggesting that intestinal ILC1 expansion and the ensuing IFN-γ-induced IDO upregulation may be the primary pathway mediating the shift to the KYN pathway in POCD. The ILC1-KYN pathway in the intestine could be a promising therapeutic target for POCD.
Collapse
Affiliation(s)
- Wan-Bing Dai
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xiao Zhang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xu-Liang Jiang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Yi-Zhe Zhang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Ling-Ke Chen
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Wei-Tian Tian
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xiao-Xin Zhou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xiao-Yu Sun
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Li-Li Huang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xi-Yao Gu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xue-Mei Chen
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xiao-Dan Wu
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian, China
| | - Jie Tian
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China.
| | - Lei Shen
- Shanghai institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiaotong University, School of Medicine, Shanghai, China.
| | - Dian-San Su
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China; Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
142
|
Tragantzopoulou P, Giannouli V. Spatial Orientation Assessment in the Elderly: A Comprehensive Review of Current Tests. Brain Sci 2024; 14:898. [PMID: 39335393 PMCID: PMC11430543 DOI: 10.3390/brainsci14090898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Spatial orientation and navigation are complex cognitive functions that integrate sensory information, attention, and memory, enabling individuals to locate themselves in their environment. These abilities decline with age, signaling cognitive impairment in neurological patients, and significantly limit the autonomy of the elderly. Current neuropsychological assessments fall short in accurately measuring everyday wayfinding abilities, particularly in borderline cases of cognitive decline. This paper reviews various neuropsychological assessments, including Benton's Judgment of Line Orientation Test, the Almeria Spatial Memory Recognition Test, the Spatial Span subtest from the Wechsler Memory Scale, and the Spatial Orientation in Immersive Virtual Environment Maze Test, evaluating their effectiveness in delineating spatial orientation and navigation skills. The review identifies significant gaps in the validity and reliability of these tests, particularly in their shortened versions, and highlights the potential of virtual reality environments as promising tools for improving diagnostic precision. The findings underscore the need for further research to refine these tools, ensuring they accurately capture cognitive decline and improve the differential diagnosis of neurodegenerative conditions like Alzheimer's disease. Such advancements hold promise for enhancing the quality of care and autonomy for the elderly.
Collapse
Affiliation(s)
| | - Vaitsa Giannouli
- School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| |
Collapse
|
143
|
Wen Y, Fu Z, Li J, Liu M, Wang X, Chen J, Chen Y, Wang H, Wen S, Zhang K, Deng Y. Targeting m 6A mRNA demethylase FTO alleviates manganese-induced cognitive memory deficits in mice. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:134969. [PMID: 38908185 DOI: 10.1016/j.jhazmat.2024.134969] [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: 03/22/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Manganese (Mn) induced learning and memory deficits through mechanisms that are not fully understood. In this study, we discovered that the demethylase FTO was significantly downregulated in hippocampal neurons in an experimental a mouse model of Mn exposure. This decreased expression of FTO was associated with Mn-induced learning and memory impairments, as well as the dysfunction in synaptic plasticity and damage to regional neurons. The overexpression of FTO, or its positive modulation with agonists, provides protection against neurological damage and cognitive impairments. Mechanistically, FTO interacts synergistically with the reader YTHDF3 to facilitate the degradation of GRIN1 and GRIN3B through the m6A modification pathway. Additionally, Mn decreases the phosphorylation of SOX2, which specifically impairs the transcriptional regulation of FTO activity. Additionally, we found that the natural compounds artemisinin and apigenin that can bind molecularly with SOX2 and reduce Mn-induced cognitive dysfunction in mice. Our findings suggest that the SOX2-FTO-Grins axis represents a viable target for addressing Mn-induced neurotoxicity and cognitive impairments.
Collapse
Affiliation(s)
- Yi Wen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Zhushan Fu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Jiashuo Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China; Department of Occupational and Environmental Health, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Mingyue Liu
- Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang, China; Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xinmiao Wang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Jingqi Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Yue Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Haocheng Wang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Sihang Wen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China
| | - Ke Zhang
- Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang, China; Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University, Shenyang, China; Engineering research center of Liaoning Province on environmental health technology and equipment, China Medical University, Shenyang, China; Institute of Health Professions Education Assessment and Reform, China Medical University, Shenyang, China.
| |
Collapse
|
144
|
Rehman Z, Alqahtani F, Ashraf W, Rasool MF, Muneeb Anjum SM, Ahmad T, Alsanea S, Alasmari F, Imran I. Neuroprotective potential of topiramate, pregabalin and lacosamide combination in a rat model of acute SE and intractable epilepsy: Perspectives from electroencephalographic, neurobehavioral and regional degenerative analysis. Eur J Pharmacol 2024; 978:176792. [PMID: 38950834 DOI: 10.1016/j.ejphar.2024.176792] [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: 03/20/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
The lithium-pilocarpine model is commonly used to recapitulate characteristics of human intractable focal epilepsy. In the current study, we explored the impact of topiramate (TPM) alone and in combination with pregabalin and lacosamide administration for 6 weeks on the evolution of spontaneous recurrent seizures (SRS) and disease-modifying potential on associated neuropsychiatric comorbidities. In addition, redox impairments and neurodegeneration in hippocampus regions vulnerable to temporal lobe epilepsy (TLE) were assessed by cresyl violet staining. Results revealed that acute electrophysiological (EEG) profiling of the ASD cocktail markedly halted sharp ictogenic spikes as well as altered dynamics of brain wave oscillations thus validating the need for polytherapy vs. monotherapy. In TLE animals, pharmacological intervention for 6 weeks with topiramate 10 mg/kg in combination with PREG and LAC at the dose of 20 mg/kg exhibited marked protection from SRS incidence, improved body weight, offensive aggression, anxiety-like behavior, cognitive impairments, and depressive-like behavior (p < 0.05). Moreover, combination therapy impeded redox impairments as evidenced by decreased MDA and AchE levels and increased activity of antioxidant SOD, GSH enzymes. Furthermore, polytherapy rescued animals from SE-induced neurodegeneration with increased neuronal density in CA1, CA3c, CA3ab, hilus, and granular cell layer (GCL) of the dentate gyrus. In conclusion, early polytherapy with topiramate in combination with pregabalin and lacosamide prompted synergy and prevented epileptogenesis with associated psychological and neuropathologic alterations.
Collapse
Affiliation(s)
- Zohabia Rehman
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Waseem Ashraf
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Syed Muhammad Muneeb Anjum
- The Institute of Pharmaceutical Sciences, University of Veterinary & Animal Sciences, Lahore, 75270, Pakistan
| | - Tanveer Ahmad
- Institut pour L'Avancée des Biosciences, Centre de Recherche UGA, INSERM U1209, CNRS 5309, Université Grenoble Alpes, France
| | - Sary Alsanea
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan.
| |
Collapse
|
145
|
Chao YW, Tung YT, Yang SC, Shirakawa H, Su LH, Loe PY, Chiu WC. The Effects of Rice Bran on Neuroinflammation and Gut Microbiota in Ovariectomized Mice Fed a Drink with Fructose. Nutrients 2024; 16:2980. [PMID: 39275295 PMCID: PMC11397027 DOI: 10.3390/nu16172980] [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: 08/13/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/16/2024] Open
Abstract
Rice bran, which is abundant in dietary fiber and phytochemicals, provides multiple health benefits. Nonetheless, its effects on neuroinflammation and gut microbiota in postmenopausal conditions are still not well understood. This study investigated the effects of rice bran and/or tea seed oil supplementation in d-galactose-injected ovariectomized (OVX) old mice fed a fructose drink. The combination of d-galactose injection, ovariectomy, and fructose drink administration creates a comprehensive model that simulates aging in females under multiple metabolic stressors, including oxidative stress, estrogen deficiency, and high-sugar diets, and allows the study of their combined impact on metabolic disorders and related diseases. Eight-week-old and 6-8-month-old female C57BL/6 mice were used. The mice were divided into six groups: a sham + young mice, a sham + old mice, an OVX + soybean oil, an OVX + soybean oil with rice bran, an OVX + tea seed oil (TO), and an OVX + TO with rice bran diet group. The OVX groups were subcutaneously injected with d-galactose (100 mg/kg/day) and received a 15% (v/v) fructose drink. The rice bran and tea seed oil supplementation formed 10% of the diet (w/w). The results showed that the rice bran with TO diet increased the number of short-chain fatty acid (SCFA)-producing Clostridia and reduced the number of endotoxin-producing Tannerellaceae, which mitigated imbalances in the gut-liver-brain axis. Rice bran supplementation reduced the relative weight of the liver, levels of hepatic triglycerides and total cholesterol; aspartate transaminase and alanine aminotransferase activity; brain levels of proinflammatory cytokines, including interleukin-1β and tumor necrosis factor-α; and plasma 8-hydroxy-2-deoxyguanosine. This study concludes that rice bran inhibits hepatic fat accumulation, which mitigates peripheral metaflammation and oxidative damage and reduces neuroinflammation in the brain.
Collapse
Affiliation(s)
- Yu-Wen Chao
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Tang Tung
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
- Advanced Plant and Food Crop Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Suh-Ching Yang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Hitoshi Shirakawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Li-Han Su
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Pei-Yu Loe
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Wan-Chun Chiu
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
- Department of Nutrition, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| |
Collapse
|
146
|
Sun Y, Zhou D, Wang Y, Wang Z, Zhang D, Qian Z, Yan J, Li Z, Huang G, Li W. Medium-chain triglycerides combined with DHA improve cognitive function by inhibiting neurocyte apoptosis of the brain in SAMP8 mice. Exp Gerontol 2024; 194:112520. [PMID: 38992823 DOI: 10.1016/j.exger.2024.112520] [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: 03/05/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Medium-chain triglycerides (MCTs) and docosahexaenoic acid (DHA, Cn-3, 22:6) are essential in improving cognitive function and protecting neurocytes. This study explored the effects of the combined intervention of MCTs and DHA on inhibiting neurocyte apoptosis of the brain and improving cognitive function in senescence-accelerated mouse-prone 8 (SAMP8). Four-month-old male SAMP8 mice were randomly divided into four treatment groups (12 mice/group): DHA, MCT, DHA + MCT, and control groups, which intervened for seven months. Twelve age-matched male senescence-accelerated mouse resistant 1 (SAMR1) was used as the natural aging group. TUNEL assay and HE staining were used to assess neurocyte apoptosis and damage in the brain of mice. Moreover, the cognitive function was analyzed using the Morris water maze (MWM) and open field (OF) tests. The results showed that the cognitive function of 11-month-old SAMP8 mice decreased with age, and further pathological examination revealed the damaged neurocyte structure, karyopyknosis, cell atrophy, and even apoptosis. MCTs combined with DHA supplementation could increase octanoic acid (C8:0), decanoic acid (C10:0), and DHA levels in the serum, inhibit neurocyte apoptosis, improve neurocyte damage, moreover delay age-related cognitive decline after seven-month treatment. Furthermore, combining MCTs and DHA was significantly more beneficial than MCTs or DHA alone. In conclusion, MCTs combined with DHA could delay cognitive decline by inhibiting neurocyte apoptosis of the brain in SAMP8 mice.
Collapse
Affiliation(s)
- Yue Sun
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Dezheng Zhou
- School of Public Health, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang 315211, China
| | - Yue Wang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Zehao Wang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Dalong Zhang
- Department of Toxicology, Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| | - Zhiyong Qian
- Department of Toxicology, Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| | - Jing Yan
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China; Department of Social Medicine and Health Administration, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Zhenshu Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Guowei Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Wen Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China.
| |
Collapse
|
147
|
Li Y, Yan Z, Shao N, Tang S, Zhang X, Liu XM, Tang J. Dual orexin receptor antagonist ameliorates sleep deprivation-induced learning and memory impairment in APP/PS1 mice. Sleep Med 2024; 121:303-314. [PMID: 39047304 DOI: 10.1016/j.sleep.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Sleep is considered closely related to cognitive function, and cognitive impairment is the main clinical manifestation of Alzheimer's disease (AD). Sleep disturbance in AD patients is more severe than that in healthy elderly individuals. Additionally, sleep deprivation reportedly increases the activity of the hypothalamic orexin system and the risk of AD. To investigate whether intervention with the orexin system can improve sleep disturbance in AD and its impact on AD pathology. In this study, six-month-old amyloid precursor protein/presenilin 1 mice were subjected to six weeks of chronic sleep deprivation and injected intraperitoneally with almorexant, a dual orexin receptor antagonist (DORA), to investigate the effects and mechanisms of sleep deprivation and almorexant intervention on learning and memory in mice with AD. We found that sleep deprivation aggravated learning and memory impairment and increased brain β-amyloid (Aβ) deposition in mice with AD. The application of almorexant can increase the total sleep time of sleep-deprived mice and reduce cognitive impairment and Aβ deposition, which is related to the improvement in Aquaporin-4 polarity. Thus, DORA may be an effective strategy for delaying the progression of AD patients by improving the sleep disturbances.
Collapse
Affiliation(s)
- Yaran Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Zian Yan
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Na Shao
- Department of Neurology, Shandong Provincial Qian Foshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shi Tang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, China.
| | - Xiao Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Xiao Min Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Jiyou Tang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, China
| |
Collapse
|
148
|
Chauhan M, Singh K, Chongtham C, A G A, Sharma P. miR-449a mediated repression of the cell cycle machinery prevents neuronal apoptosis. J Biol Chem 2024; 300:107698. [PMID: 39173945 PMCID: PMC11419829 DOI: 10.1016/j.jbc.2024.107698] [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: 04/27/2024] [Revised: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 08/24/2024] Open
Abstract
Aberrant activation of the cell cycle of terminally differentiated neurons results in their apoptosis and is known to contribute to neuronal loss in various neurodegenerative disorders like Alzheimer's Disease. However, the mechanisms that regulate cell cycle-related neuronal apoptosis are poorly understood. We identified several miRNA that are dysregulated in neurons from a transgenic APP/PS1 mouse model for AD (TgAD). Several of these miRNA are known to and/or are predicted to target cell cycle-related genes. Detailed investigation on miR-449a revealed the following: a, it promotes neuronal differentiation by suppressing the neuronal cell cycle; b, its expression in cortical neurons was impaired in response to amyloid peptide Aβ42; c, loss of its expression resulted in aberrant activation of the cell cycle leading to apoptosis. miR-449a may prevent cell cycle-related neuronal apoptosis by targeting cyclin D1 and protein phosphatase CDC25A, which are important for G1-S transition. Importantly, the lentiviral-mediated delivery of miR-449a in TgAD mouse brain significantly reverted the defects in learning and memory, which are associated with AD.
Collapse
Affiliation(s)
- Monika Chauhan
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India.
| | - Komal Singh
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - Chen Chongtham
- Molecular Genetics Laboratory, National Institute of Immunology, New Delhi, India
| | - Aneeshkumar A G
- Molecular Genetics Laboratory, National Institute of Immunology, New Delhi, India
| | - Pushkar Sharma
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India.
| |
Collapse
|
149
|
Penman SL, Roeder NM, Wang J, Richardson BJ, Freeman-Striegel L, Krayevsky A, Eiden RD, Chakraborty S, Thanos PK. Vaporized Δ9-tetrahydrocannabinol exposure in utero has negative effects on attention in a dose- and sex-dependent manner. Pharmacol Biochem Behav 2024; 242:173808. [PMID: 38914267 DOI: 10.1016/j.pbb.2024.173808] [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: 04/29/2024] [Revised: 06/03/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024]
Abstract
There has been an increasing use of cannabis during pregnancy in recent years. Studies have indicated that THC exposure in utero may increase the risk of attention deficits and memory impairments in adolescence. The goal of the present study is to investigate the effects of vaporized THC exposure during pregnancy on offspring memory and attention performance in early and late adolescence. Pregnant dams were exposed to vaporized THC (10 mg or 40 mg) daily from gestational day 2 until labor. Pups were given either a standard or a high-fat diet at weaning and tested in early and late adolescence in two memory tests, the Novel Object Recognition (NOR) test and the Morris Water Maze (MWM) test, and a test of attention, the Object-Based Attention (OBA) test. Rats exposed to low-dose THC showed significantly decreased object exploration in both the NOR and OBA tests, indicating decreased attention. Object exploration time in OBA was significantly lower in females than males. Additionally, post hoc analysis of MWM tests showed some differences in learning patterns for HD THC offspring in early adolescence, possibly due to diet interaction, but ultimate performance was not impacted. While there are existing studies examining prenatal exposure to THC in rodents, this is the first to our knowledge examining memory and attention in adolescence following vaporized THC exposure in utero, and we find indications that prenatal THC exposure may lead to attention deficits and altered memory performance.
Collapse
Affiliation(s)
- Samantha L Penman
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Nicole M Roeder
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Department of Psychology, University at Buffalo, Buffalo, NY, USA
| | - Jia Wang
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Brittany J Richardson
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Lily Freeman-Striegel
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Alexis Krayevsky
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Rina D Eiden
- Department of Psychology and Social Science Research Institute, The Pennsylvania State University, University Park, PA, USA
| | - Saptarshi Chakraborty
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Department of Psychology, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
150
|
He X, Peng Y, Huang S, Xiao Z, Li G, Zuo Z, Zhang L, Shuai X, Zheng H, Hu X. Blood Brain Barrier-Crossing Delivery of Felodipine Nanodrug Ameliorates Anxiety-Like Behavior and Cognitive Impairment in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401731. [PMID: 38981028 PMCID: PMC11425895 DOI: 10.1002/advs.202401731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/29/2024] [Indexed: 07/11/2024]
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder leading to cognitive decline. Excessive cytosolic calcium (Ca2+) accumulation plays a critical role in the pathogenesis of AD since it activates the NOD-like receptor family, pyrin domain containing 3 (NLRP3), switches the endoplasmic reticulum (ER) unfolded protein response (UPR) toward proapoptotic signaling and promotes Aβ seeding. Herein, a liposomal nanodrug (felodipine@LND) is developed incorporating a calcium channel antagonist felodipine for Alzheimer's disease treatment through a low-intensity pulse ultrasound (LIPUS) irradiation-assisted blood brain barrier (BBB)-crossing drug delivery. The multifunctional felodipine@LND is effectively delivered to diseased brain through applying a LIPUS irradiation to the skull, which resulted in a series of positive effects against AD. Markedly, the nanodrug treatment switched the ER UPR toward antioxidant signaling, prevented the surface translocation of ER calreticulin (CALR) in microglia, and inhibited the NLRP3 activation and Aβ seeding. In addition, it promoted the degradation of damaged mitochondria via mitophagy, thereby inhibiting the neuronal apoptosis. Therefore, the anxiety-like behavior and cognitive impairment of 5xFAD mice with AD is significantly ameliorated, which manifested the potential of LIPUS - assisted BBB-crossing delivery of felodipine@LND to serve as a paradigm for AD therapy based on the well-recognized clinically available felodipine.
Collapse
Affiliation(s)
- Xiaofei He
- Department of Rehabilitation MedicineThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| | - Yuan Peng
- Department of Rehabilitation MedicineGuangzhou First People's HospitalGuangzhou510180China
| | - Sicong Huang
- School of Materials Science and Engineering Sun Yat‐sen UniversityGuangzhou510275China
| | - Zecong Xiao
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Ge Li
- Guangdong Provincial Key Laboratory of Laboratory AnimalsGuangdong Laboratory Animals Monitoring Institute11 Fengxin RoadGuangzhouGuangdong510663China
| | - Zejie Zuo
- Department of Rehabilitation MedicineThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| | - Liying Zhang
- Department of Rehabilitation MedicineThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| | - Xintao Shuai
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Haiqing Zheng
- Department of Rehabilitation MedicineThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| | - Xiquan Hu
- Department of Rehabilitation MedicineThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| |
Collapse
|