1
|
Zhang Y, Chen F, Li X, Xu Y, Liu X, Barkat MQ, Choudhary MI, Chang Q, Jiang N. Gastrodia elata, Polygonatum sibiricum, and Poria cocos as a Functional Food Formula: Cognitive Enhancement via Modulation of Hippocampal Neuroinflammation and Neuroprotection in Sleep-Restricted Mice. Foods 2025; 14:1103. [PMID: 40238194 PMCID: PMC11988919 DOI: 10.3390/foods14071103] [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: 02/06/2025] [Revised: 03/03/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Gastrodia elata, Polygonatum sibiricum, and Poria cocos are traditional Chinese herbs commonly used as both medicinal and food ingredients, traditionally believed to improve liver and kidney functions, replenish vital energy (qi) and blood, and mitigate stress-induced damage. These herbs are combined in the Compound Gastrodia elata Formula (CGEF), a functional food formulation. Amidst growing interest in functional foods, this study explores the cognitive-enhancing effects of CGEF, focusing on cognitive function improvement. Cognitive impairment was induced in ICR mice via chronic sleep restriction. Behavioral assessments including the Y-maze test, object recognition test, Morris water maze test, and Passive avoidance test, were conducted to evaluate CGEF's effects. Serum levels of inflammatory markers and oxidative stress were quantified while in rat hippocampus tissue expressions of inflammatory, apoptotic, and neuroprotective-related protein markers were analyzed by Western blotting. Neurotransmitter concentrations in both the hippocampus and prefrontal cortex were determined by LC-MS/MS. CGEF significantly alleviated cognitive impairments across all behavioral tests. The underlying mechanisms likely involve a reduction in oxidative stress and peripheral inflammatory factors, and suppression of the TLR2/MyD88/NF-κB signaling cascade in the hippocampus, thereby mitigating neuroinflammation and neuronal apoptosis. Furthermore, CGEF modulated the PI3K/AKT/GSK3β signaling pathway, potentially contributing to neuronal integrity and synaptic plasticity maintenance. CGEF also restored neurotransmitter balance and regulated tryptophan metabolism, further alleviating cognitive deficits associated with sleep disruption. These findings suggest CGEF's potential as a functional food for reversing cognitive impairments caused by chronic sleep restriction, primarily through its anti-inflammatory and neuroprotective effects.
Collapse
Affiliation(s)
- Yiwen Zhang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Fang Chen
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Xueyan Li
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yanfei Xu
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Xinmin Liu
- Sino-Pakistan Center on Traditional Chinese Medicine, Hunan University of Medicine, Huaihua 418000, China
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | | | - Muhammad Iqbal Choudhary
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Qi Chang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Ning Jiang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| |
Collapse
|
2
|
Riffo-Lepe N, González-Sanmiguel J, Armijo-Weingart L, Saavedra-Sieyes P, Hernandez D, Ramos G, San Martín LS, Aguayo LG. Synaptic and synchronic impairments in subcortical brain regions associated with early non-cognitive dysfunction in Alzheimer's disease. Neural Regen Res 2025; 21:01300535-990000000-00688. [PMID: 39885666 PMCID: PMC12094569 DOI: 10.4103/nrr.nrr-d-24-01052] [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: 09/08/2024] [Revised: 11/16/2024] [Accepted: 12/21/2024] [Indexed: 02/01/2025] Open
Abstract
ABSTRACT For many decades, Alzheimer's disease research has primarily focused on impairments within cortical and hippocampal regions, which are thought to be related to cognitive dysfunctions such as memory and language deficits. The exact cause of Alzheimer's disease is still under debate, making it challenging to establish an effective therapy or early diagnosis. It is widely accepted that the accumulation of amyloid-beta peptide in the brain parenchyma leads to synaptic dysfunction, a critical step in Alzheimer's disease development. The traditional amyloid cascade model is initiated by accumulating extracellular amyloid-beta in brain areas essential for memory and language. However, while it is possible to reduce the presence of amyloid-beta plaques in the brain with newer immunotherapies, cognitive symptoms do not necessarily improve. Interestingly, recent studies support the notion that early alterations in subcortical brain regions also contribute to brain damage and precognitive decline in Alzheimer's disease. A body of recent evidence suggests that early Alzheimer's disease is associated with alterations (e.g., motivation, anxiety, and motor impairment) in subcortical areas, such as the striatum and amygdala, in both human and animal models. Also, recent data indicate that intracellular amyloid-beta appears early in subcortical regions such as the nucleus accumbens, locus coeruleus, and raphe nucleus, even without extracellular amyloid plaques. The reported effects are mainly excitatory, increasing glutamatergic transmission and neuronal excitability. In agreement, data in Alzheimer's disease patients and animal models show an increase in neuronal synchronization that leads to electroencephalogram disturbances and epilepsy. The data indicate that early subcortical brain dysfunctions might be associated with non-cognitive symptoms such as anxiety, irritability, and motivation deficits, which precede memory loss and language alterations. Overall, the evidence reviewed suggests that subcortical brain regions could explain early dysfunctions and perhaps be targets for therapies to slow disease progression. Future research should focus on these non-traditional brain regions to reveal early pathological alterations and underlying mechanisms to advance our understanding of Alzheimer's disease beyond the traditionally studied hippocampal and cortical circuits.
Collapse
Affiliation(s)
- Nicolás Riffo-Lepe
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Juliana González-Sanmiguel
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Lorena Armijo-Weingart
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Concepción, Chile
| | - Paulina Saavedra-Sieyes
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - David Hernandez
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Gerson Ramos
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Loreto S. San Martín
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| | - Luis G. Aguayo
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| |
Collapse
|
3
|
Zhao K, Wang D, Wang D, Chen P, Wei Y, Tu L, Chen Y, Tang Y, Yao H, Zhou B, Lu J, Wang P, Liao Z, Chen Y, Han Y, Zhang X, Liu Y. Macroscale connectome topographical structure reveals the biomechanisms of brain dysfunction in Alzheimer's disease. SCIENCE ADVANCES 2024; 10:eado8837. [PMID: 39392880 PMCID: PMC11809497 DOI: 10.1126/sciadv.ado8837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 09/11/2024] [Indexed: 10/13/2024]
Abstract
The intricate spatial configurations of brain networks offer essential insights into understanding the specific patterns of brain abnormalities and the underlying biological mechanisms associated with Alzheimer's disease (AD), normal aging, and other neurodegenerative disorders. This study investigated alterations in the topographical structure of the brain related to aging and neurodegenerative diseases by analyzing brain gradients derived from structural MRI data across multiple cohorts (n = 7323). The analysis identified distinct gradient patterns in AD, aging, and other neurodegenerative conditions. Gene enrichment analysis indicated that inorganic ion transmembrane transport was the most significant term in normal aging, while chemical synaptic transmission is a common enrichment term across various neurodegenerative diseases. Moreover, the findings show that each disorder exhibits unique dysfunctional neurophysiological characteristics. These insights are pivotal for elucidating the distinct biological mechanisms underlying AD, thereby enhancing our understanding of its unique clinical phenotypes in contrast to normal aging and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Kun Zhao
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
- Research Institute of Shandong University: Magnetic Field-free Medicine & Functional Imaging, Jinan, China
- Shandong Key Laboratory: Magnetic Field-free Medicine & Functional Imaging (MF), Jinan, China
| | - Dong Wang
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Pindong Chen
- School of Artificial Intelligence, University of Chinese Academy of Sciences & Brainnetome Center, Chinese Academy of Sciences, Beijing, China
| | - Yongbin Wei
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Liyun Tu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yuqi Chen
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yi Tang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hongxiang Yao
- Department of Radiology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Bo Zhou
- Department of Neurology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Jie Lu
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Pan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhengluan Liao
- Department of Psychiatry, People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Yan Chen
- Department of Psychiatry, People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
- Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Xi Zhang
- Department of Neurology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences & Brainnetome Center, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
4
|
Zhang S, Lu J, Jin Z, Xu H, Zhang D, Chen J, Wang J. Gut microbiota metabolites: potential therapeutic targets for Alzheimer's disease? Front Pharmacol 2024; 15:1459655. [PMID: 39355779 PMCID: PMC11442227 DOI: 10.3389/fphar.2024.1459655] [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: 07/04/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive decline in cognitive function, which significantly increases pain and social burden. However, few therapeutic interventions are effective in preventing or mitigating the progression of AD. An increasing number of recent studies support the hypothesis that the gut microbiome and its metabolites may be associated with upstream regulators of AD pathology. Methods In this review, we comprehensively explore the potential mechanisms and currently available interventions targeting the microbiome for the improvement of AD. Our discussion is structured around modern research advancements in AD, the bidirectional communication between the gut and brain, the multi-target regulatory effects of microbial metabolites on AD, and therapeutic strategies aimed at modulating gut microbiota to manage AD. Results The gut microbiota plays a crucial role in the pathogenesis of AD through continuous bidirectional communication via the microbiota-gut-brain axis. Among these, microbial metabolites such as lipids, amino acids, bile acids and neurotransmitters, especially sphingolipids and phospholipids, may serve as central components of the gut-brain axis, regulating AD-related pathogenic mechanisms including β-amyloid metabolism, Tau protein phosphorylation, and neuroinflammation. Additionally, interventions such as probiotic administration, fecal microbiota transplantation, and antibiotic use have also provided evidence supporting the association between gut microbiota and AD. At the same time, we propose an innovative strategy for treating AD: a healthy lifestyle combined with targeted probiotics and other potential therapeutic interventions, aiming to restore intestinal ecology and microbiota balance. Conclusion Despite previous efforts, the molecular mechanisms by which gut microbes act on AD have yet to be fully described. However, intestinal microorganisms may become an essential target for connecting the gut-brain axis and improving the symptoms of AD. At the same time, it requires joint exploration by multiple centers and multiple disciplines.
Collapse
Affiliation(s)
- Shanshan Zhang
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Ziqi Jin
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Hanying Xu
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Dongmei Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Jianan Chen
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jian Wang
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
5
|
Yang X, Wang Z, Li H, Qin W, Liu N, Liu Z, Wang S, Xu J, Wang J, for the Alzheimer's Disease Neuroimaging Initiative. Polygenic Score for Conscientiousness Is a Protective Factor for Reversion from Mild Cognitive Impairment to Normal Cognition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309889. [PMID: 38838096 PMCID: PMC11304237 DOI: 10.1002/advs.202309889] [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: 12/16/2023] [Revised: 05/21/2024] [Indexed: 06/07/2024]
Abstract
Spontaneous reversion from mild cognitive impairment (MCI) to normal cognition (NC) is little known. Based on the data of the Genetics of Personality Consortium and MCI participants from Alzheimer's Disease Neuroimaging Initiative, the authors investigate the effect of polygenic scores (PGS) for personality traits on the reversion of MCI to NC and its underlying neurobiology. PGS analysis reveals that PGS for conscientiousness (PGS-C) is a protective factor that supports the reversion from MCI to NC. Gene ontology enrichment analysis and tissue-specific enrichment analysis indicate that the protective effect of PGS-C may be attributed to affecting the glutamatergic synapses of subcortical structures, such as hippocampus, amygdala, nucleus accumbens, and caudate nucleus. The structural covariance network (SCN) analysis suggests that the left whole hippocampus and its subfields, and the left whole amygdala and its subnuclei show significantly stronger covariance with several high-cognition relevant brain regions in the MCI reverters compared to the stable MCI participants, which may help illustrate the underlying neural mechanism of the protective effect of PGS-C.
Collapse
Affiliation(s)
- Xuan Yang
- Department of RadiologyTianjin Key Lab of Functional Imaging & Tianjin Institute of RadiologyTianjin Medical University General HospitalTianjin300052P. R. China
- Department of RadiologyJining No.1 People's HospitalJiningShandong272000P. R. China
| | - Zirui Wang
- Department of RadiologyTianjin Key Lab of Functional Imaging & Tianjin Institute of RadiologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Haonan Li
- Department of RadiologyTianjin Key Lab of Functional Imaging & Tianjin Institute of RadiologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Wen Qin
- Department of RadiologyTianjin Key Lab of Functional Imaging & Tianjin Institute of RadiologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Nana Liu
- Department of RadiologyTianjin Key Lab of Functional Imaging & Tianjin Institute of RadiologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Zhixuan Liu
- Department of RadiologyTianjin Key Lab of Functional Imaging & Tianjin Institute of RadiologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Siqi Wang
- Department of RadiologyTianjin Key Lab of Functional Imaging & Tianjin Institute of RadiologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Jiayuan Xu
- Department of RadiologyTianjin Key Lab of Functional Imaging & Tianjin Institute of RadiologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Junping Wang
- Department of RadiologyTianjin Key Lab of Functional Imaging & Tianjin Institute of RadiologyTianjin Medical University General HospitalTianjin300052P. R. China
| | | |
Collapse
|
6
|
Wu X, Zhu X, Pan Y, Gu X, Liu X, Chen S, Zhang Y, Xu T, Xu N, Sun S. Amygdala neuronal dyshomeostasis via 5-HT receptors mediates mood and cognitive defects in Alzheimer's disease. Aging Cell 2024; 23:e14187. [PMID: 38716507 PMCID: PMC11320345 DOI: 10.1111/acel.14187] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 08/15/2024] Open
Abstract
Behavioral changes or neuropsychiatric symptoms (NPSs) are common features in dementia and are associated with accelerated cognitive impairment and earlier deaths. However, how NPSs are intertwined with cognitive decline remains elusive. In this study, we identify that the basolateral amygdala (BLA) is a key brain region that is associated with mood disorders and memory decline in the AD course. During the process from pre- to post-onset in AD, the dysfunction of parvalbumin (PV) interneurons and pyramidal neurons in the amygdala leads to hyperactivity of pyramidal neurons in the basal state and insensitivity to external stimuli. We further demonstrate that serotonin (5-HT) receptors in distinct neurons synergistically regulate the BLA microcircuit of AD rather than 5-HT levels, in which both restrained inhibitory inputs by excessive 5-HT1AR signaling in PV interneurons and depolarized pyramidal neurons via upregulated 5-HT2AR contribute to aberrant neuronal hyperactivity. Downregulation of these two 5-HT receptors simultaneously enables neurons to resist β-amyloid peptides (Aβ) neurotoxicity and ameliorates the mood and cognitive defects. Therefore, our study reveals a crucial role of 5-HT receptors for regulating neuronal homeostasis in AD pathogenesis, and this would provide early intervention and potential targets for AD cognitive decline.
Collapse
Affiliation(s)
- Xin‐Rong Wu
- Department of NeurologyInstitute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiao‐Na Zhu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuan‐Bo Pan
- Department of Neurosurgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue Gu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xian‐Dong Liu
- Department of NeurologyInstitute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Si Chen
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yu Zhang
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tian‐Le Xu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Nan‐Jie Xu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
- Songjiang Hospital and Songjiang Research InstituteShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Emotions and Affective DisordersShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Suya Sun
- Department of NeurologyInstitute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|