1
|
Chen L, Zhuang Z, Duan H, Lv D, Hong S, Chen P, He B, Shen Z. Corilagin improves cognitive impairment in APP/PS1 mice by reducing Aβ generation and enhancing synaptic plasticity. Eur J Pharmacol 2024; 981:176893. [PMID: 39134295 DOI: 10.1016/j.ejphar.2024.176893] [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: 11/09/2023] [Revised: 02/24/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
Alzheimer's disease (AD) is closely associated with the neurotoxic effects of amyloid-β (Aβ), leading to synaptic damage, neuronal loss and cognitive dysfunction. Previous in vitro studies have demonstrated the potential of corilagin to counteract Aβ-induced oxidative stress, inflammatory injury, and β-site amyloid precursor protein cleaving enzyme-1 (BACE1) activity in Aβ production. However, the in vivo protective effects of corilagin on Alzheimer's disease remain unexplored. The purpose of this study was to investigate the protective effects of corilagin on APP/PS1 mice and the underlying mechanisms. The cognitive function of the mice was assessed by step-through passive avoidance and Morris water maze tests. Nissl staining was used to evaluate neuronal damage in the hippocampus. ELISA and Western blotting analyses were used to determine the associated protein expression. Transmission electron microscopy was utilized to observe the synaptic ultrastructure of hippocampal neurons. Golgi staining was applied to assess dendritic morphology and dendritic spine density in hippocampal pyramidal neurons. Immunohistochemistry and Western blotting were performed to examine the expression of synaptic-associated proteins. The results showed that corilagin improves learning and memory in APP/PS1 mice, reduces hippocampal neuron damage, inhibits BACE1 and reduces Aβ generation. It also improves synaptic plasticity and the expression of synaptic-associated proteins. Corilagin effectively reduces Aβ generation by inhibiting BACE1, ultimately reducing neuronal loss and enhancing synaptic plasticity to improve synaptic transmission. This study sheds light on the potential therapeutic role of corilagin in Alzheimer's disease.
Collapse
Affiliation(s)
- Linyi Chen
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China
| | - Zhujun Zhuang
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China
| | - Hengqian Duan
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China
| | - Di Lv
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China
| | - Shengxiong Hong
- Laboratory Animal Department, Kunming Medical University, Kunming, 650031, Yunnan, China
| | - Peng Chen
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China.
| | - Bo He
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China.
| | - Zhiqiang Shen
- School of Pharmaceutical Sciences, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China.
| |
Collapse
|
2
|
Sung KC, Wang LY, Wang CC, Chu CH, Sun HS, Hsiao YH. Enhanced hippocampal TIAM2S expression alleviates cognitive deficits in Alzheimer's disease model mice. Pharmacol Rep 2024; 76:1032-1043. [PMID: 39012419 DOI: 10.1007/s43440-024-00623-3] [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: 12/21/2023] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Dendritic spine dysfunction is a key feature of Alzheimer's disease (AD) pathogenesis. Human T-cell lymphoma invasion and metastasis 2 (TIAM2) is expressed in two isoforms, the full length (TIAM2L) and a short transcript (TIAM2S). Compared to TIAM2L protein, which is undetectable, TIAM2S protein is abundant in human brain tissue, especially the hippocampus, and can promote neurite outgrowth in our previous findings. However, whether enhanced hippocampal TIAM2S expression can alleviate cognitive deficits in Alzheimer's disease model mice remains unclear. METHODS We crossbred 3xTg-AD with TIAM2S mice to generate an AD mouse model that carries the human TIAM2S gene (3xTg-AD/TIAM2S mice). The Morris water maze and object location tests assessed hippocampus-dependent spatial memory. Lentiviral-driven shRNA or cDNA approaches were used to manipulate hippocampal TIAM2S expression. Golgi staining and Sholl analysis were utilized to measure neuronal dendrites and dendritic spines in the mouse hippocampi. RESULTS Compared to 3xTg-AD mice, 3xTg-AD/TIAM2S mice displayed improved cognitive functions. According to the hippocampus is one of the earliest affected brain regions by AD, we further injected TIAM2S shRNA or TIAM2S cDNA into mouse hippocampi to confirm whether manipulating hippocampal TIAM2S expression could affect AD-related cognitive functions. The results showed that the reduced hippocampal TIAM2S expression in 3xTg-AD/TIAM2S mice abolished the memory improvement effect, whereas increased hippocampal TIAM2S levels alleviated cognitive deficits in 3xTg-AD mice. Furthermore, we found that TIAM2S-mediated memory improvement was achieved by regulating dendritic plasticity. CONCLUSIONS These results will provide new insights into connecting TIAM2S with AD and support the notion that TIAM2S should be investigated as potential AD therapeutic targets.
Collapse
Affiliation(s)
- Kuan-Chin Sung
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Li-Yun Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Che-Chuan Wang
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Chun-Hsien Chu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - H Sunny Sun
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Hsin Hsiao
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
3
|
Hu NW, Ondrejcak T, Klyubin I, Yang Y, Walsh DM, Livesey FJ, Rowan MJ. Patient-derived tau and amyloid-β facilitate long-term depression in vivo: role of tumour necrosis factor-α and the integrated stress response. Brain Commun 2024; 6:fcae333. [PMID: 39391333 PMCID: PMC11465085 DOI: 10.1093/braincomms/fcae333] [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: 01/23/2024] [Revised: 08/22/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease is characterized by a progressive cognitive decline in older individuals accompanied by the deposition of two pathognomonic proteins amyloid-β and tau. It is well documented that synaptotoxic soluble amyloid-β aggregates facilitate synaptic long-term depression, a major form of synaptic weakening that correlates with cognitive status in Alzheimer's disease. Whether synaptotoxic tau, which is also associated strongly with progressive cognitive decline in patients with Alzheimer's disease and other tauopathies, also causes facilitation remains to be clarified. Young male adult and middle-aged rats were employed. Synaptotoxic tau and amyloid-β were obtained from different sources including (i) aqueous brain extracts from patients with Alzheimer's disease and Pick's disease tauopathy; (ii) the secretomes of induced pluripotent stem cell-derived neurons from individuals with trisomy of chromosome 21; and (iii) synthetic amyloid-β. In vivo electrophysiology was performed in urethane anaesthetized animals. Evoked field excitatory postsynaptic potentials were recorded from the stratum radiatum in the CA1 area of the hippocampus with electrical stimulation to the Schaffer collateral-commissural pathway. To study the enhancement of long-term depression, relatively weak low-frequency electrical stimulation was used to trigger peri-threshold long-term depression. Synaptotoxic forms of tau or amyloid-β were administered intracerebroventricularly. The ability of agents that inhibit the cytokine tumour necrosis factor-α or the integrated stress response to prevent the effects of amyloid-β or tau on long-term depression was assessed after local or systemic injection, respectively. We found that diffusible tau from Alzheimer's disease or Pick's disease patients' brain aqueous extracts or the secretomes of trisomy of chromosome 21 induced pluripotent stem cell-derived neurons, like Alzheimer's disease brain-derived amyloid-β and synthetic oligomeric amyloid-β, potently enhanced synaptic long-term depression in live rats. We further demonstrated that long-term depression facilitation by both tau and amyloid-β was age-dependent, being more potent in middle-aged compared with young animals. Finally, at the cellular level, we provide pharmacological evidence that tumour necrosis factor-α and the integrated stress response are downstream mediators of long-term depression facilitation by both synaptotoxic tau and amyloid-β. Overall, these findings reveal the promotion of an age-dependent synaptic weakening by both synaptotoxic tau and amyloid-β. Pharmacologically targeting shared mechanisms of tau and amyloid-β synaptotoxicity, such as tumour necrosis factor-α or the integrated stress response, provides an attractive strategy to treat early Alzheimer's disease.
Collapse
Affiliation(s)
- Neng-Wei Hu
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Dublin, Ireland
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Tomas Ondrejcak
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Dublin, Ireland
| | - Igor Klyubin
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Dublin, Ireland
| | - Yin Yang
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Dublin, Ireland
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Frederick J Livesey
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, London WC1N 1DZ, UK
| | - Michael J Rowan
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Dublin, Ireland
| |
Collapse
|
4
|
Ray A, Loghinov I, Ravindranath V, Barth AL. Early hippocampal hyperexcitability and synaptic reorganization in mouse models of amyloidosis. iScience 2024; 27:110629. [PMID: 39262788 PMCID: PMC11388185 DOI: 10.1016/j.isci.2024.110629] [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: 01/30/2024] [Revised: 05/09/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
The limited success of plaque-reducing therapies in Alzheimer's disease suggests that early treatment might be more effective in delaying or reversing memory impairments. Toward this end, it is important to establish the progression of synaptic and circuit changes before onset of plaques or cognitive deficits. Here, we used quantitative, fluorescence-based methods for synapse detection in CA1 pyramidal neurons to investigate the interaction between abnormal circuit activity, measured by Fos-immunoreactivity, and synapse reorganization in mouse models of amyloidosis. Using a genetically encoded, fluorescently labeled synaptic marker in juvenile mice (prior to sexual maturity), we find both synapse gain and loss depending on dendritic location. This progresses to broad synapse loss in aged mice. Elevated hippocampal activity in both CA3 and CA1 was present at weaning and preceded this reorganization. Thus, Aβ overproduction may initiate abnormal activity and subsequent input-specific synapse plasticity. These findings indicate that sustained amyloidosis drives heterogeneous and progressive circuit-wide abnormalities.
Collapse
Affiliation(s)
- Ajit Ray
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Iulia Loghinov
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Vijayalakshmi Ravindranath
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, Karnataka 560012, India
- Centre for Brain Research, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Alison L Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
5
|
Liang J, Menon A, Tomco T, Bhattarai N, Smith IN, Khrestian M, Formica SV, Eng C, Buck M, Bekris LM. A Computational Approach in the Systematic Search of the Interaction Partners of Alternatively Spliced TREM2 Isoforms. Int J Mol Sci 2024; 25:9667. [PMID: 39273614 PMCID: PMC11395018 DOI: 10.3390/ijms25179667] [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: 08/05/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Alzheimer's disease is the most common form of dementia, characterized by the pathological accumulation of amyloid-beta (Aβ) plaques and tau neurofibrillary tangles. Triggering receptor expressed on myeloid cells 2 (TREM2) is increasingly recognized as playing a central role in Aβ clearance and microglia activation in AD. The TREM2 gene transcriptional product is alternatively spliced to produce three different protein isoforms. The canonical TREM2 isoform binds to DAP12 to activate downstream pathways. However, little is known about the function or interaction partners of the alternative TREM2 isoforms. The present study utilized a computational approach in a systematic search for new interaction partners of the TREM2 isoforms by integrating several state-of-the-art structural bioinformatics tools from initial large-scale screening to one-on-one corroborative modeling and eventual all-atom visualization. CD9, a cell surface glycoprotein involved in cell-cell adhesion and migration, was identified as a new interaction partner for two TREM2 isoforms, and CALM, a calcium-binding protein involved in calcium signaling, was identified as an interaction partner for a third TREM2 isoform, highlighting the potential role of cell adhesion and calcium regulation in AD.
Collapse
Affiliation(s)
- Junyi Liang
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Aditya Menon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Taylor Tomco
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Nisha Bhattarai
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Iris Nira Smith
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Maria Khrestian
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Shane V Formica
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Charis Eng
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Lynn M Bekris
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
6
|
Zhao F, Ma X, Yao B, Lu Q, Chen L. scaDA: A novel statistical method for differential analysis of single-cell chromatin accessibility sequencing data. PLoS Comput Biol 2024; 20:e1011854. [PMID: 39093856 PMCID: PMC11324137 DOI: 10.1371/journal.pcbi.1011854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/14/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024] Open
Abstract
Single-cell ATAC-seq sequencing data (scATAC-seq) has been widely used to investigate chromatin accessibility on the single-cell level. One important application of scATAC-seq data analysis is differential chromatin accessibility (DA) analysis. However, the data characteristics of scATAC-seq such as excessive zeros and large variability of chromatin accessibility across cells impose a unique challenge for DA analysis. Existing statistical methods focus on detecting the mean difference of the chromatin accessible regions while overlooking the distribution difference. Motivated by real data exploration that distribution difference exists among cell types, we introduce a novel composite statistical test named "scaDA", which is based on zero-inflated negative binomial model (ZINB), for performing differential distribution analysis of chromatin accessibility by jointly testing the abundance, prevalence and dispersion simultaneously. Benefiting from both dispersion shrinkage and iterative refinement of mean and prevalence parameter estimates, scaDA demonstrates its superiority to both ZINB-based likelihood ratio tests and published methods by achieving the highest power and best FDR control in a comprehensive simulation study. In addition to demonstrating the highest power in three real sc-multiome data analyses, scaDA successfully identifies differentially accessible regions in microglia from sc-multiome data for an Alzheimer's disease (AD) study that are most enriched in GO terms related to neurogenesis and the clinical phenotype of AD, and AD-associated GWAS SNPs.
Collapse
Affiliation(s)
- Fengdi Zhao
- Department of Biostatistics, University of Florida, Gainesville, Florida, United States of America
| | - Xin Ma
- Department of Biostatistics, University of Florida, Gainesville, Florida, United States of America
| | - Bing Yao
- Department of Human Genetics, Emory University, Atlanta, Georgia, United States of America
| | - Qing Lu
- Department of Biostatistics, University of Florida, Gainesville, Florida, United States of America
| | - Li Chen
- Department of Biostatistics, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
7
|
Nguyen HD, Kim WK, Huong Vu G. Molecular mechanisms implicated in protein changes in the Alzheimer's disease human hippocampus. Mech Ageing Dev 2024; 219:111930. [PMID: 38554950 DOI: 10.1016/j.mad.2024.111930] [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: 12/03/2023] [Revised: 01/18/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
This study aimed to elucidate the specific biochemical pathways linked to changes in proteins in the Alzheimer's disease (AD) human hippocampus. Our data demonstrate a constant rise in the expression of four proteins (VGF, GFAP, HSPB1, and APP) across all eleven studies. Notably, UBC was the most centrally involved and had increased expression in the hippocampus tissue of individuals with AD. Modified proteins in the hippocampal tissue were found to activate the innate immune system and disrupt communication across chemical synapses. Four hub proteins (CD44, APP, ITGB2, and APOE) are connected to amyloid plaques, whereas two hub proteins (RPL24 and RPS23) are related to neurofibrillary tangles (NFTs). The presence of modified proteins was discovered to trigger the activation of microglia and decrease the functioning of ribosomes and mitochondria in the hippocampus. Three significant microRNAs (hsa-miR-106b-5p, hsa-miR-17-5p, and hsa-miR-16-5p) and transcription factors (MYT1L, PIN1, and CSRNP3) have been discovered to improve our understanding of the alterations in proteins within the hippocampal tissues that lead to the progression of AD. These findings establish a path for possible treatments for AD to employ therapeutic strategies that specifically focus on the proteins or processes linked to the illness.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea; Division of Microbiology, Tulane National Primate Research Center, Tulane University, Louisiana, USA.
| | - Woong-Ki Kim
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Louisiana, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Giang Huong Vu
- Department of Public Health, Hong Bang Health Center, Hai Phong, Vietnam
| |
Collapse
|
8
|
Sutton NM, Gutiérrez-Guzmán BE, Dannenberg H, Ascoli GA. A Continuous Attractor Model with Realistic Neural and Synaptic Properties Quantitatively Reproduces Grid Cell Physiology. Int J Mol Sci 2024; 25:6059. [PMID: 38892248 PMCID: PMC11173171 DOI: 10.3390/ijms25116059] [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: 04/29/2024] [Revised: 05/25/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Computational simulations with data-driven physiological detail can foster a deeper understanding of the neural mechanisms involved in cognition. Here, we utilize the wealth of cellular properties from Hippocampome.org to study neural mechanisms of spatial coding with a spiking continuous attractor network model of medial entorhinal cortex circuit activity. The primary goal is to investigate if adding such realistic constraints could produce firing patterns similar to those measured in real neurons. Biological characteristics included in the work are excitability, connectivity, and synaptic signaling of neuron types defined primarily by their axonal and dendritic morphologies. We investigate the spiking dynamics in specific neuron types and the synaptic activities between groups of neurons. Modeling the rodent hippocampal formation keeps the simulations to a computationally reasonable scale while also anchoring the parameters and results to experimental measurements. Our model generates grid cell activity that well matches the spacing, size, and firing rates of grid fields recorded in live behaving animals from both published datasets and new experiments performed for this study. Our simulations also recreate different scales of those properties, e.g., small and large, as found along the dorsoventral axis of the medial entorhinal cortex. Computational exploration of neuronal and synaptic model parameters reveals that a broad range of neural properties produce grid fields in the simulation. These results demonstrate that the continuous attractor network model of grid cells is compatible with a spiking neural network implementation sourcing data-driven biophysical and anatomical parameters from Hippocampome.org. The software (version 1.0) is released as open source to enable broad community reuse and encourage novel applications.
Collapse
Affiliation(s)
- Nate M. Sutton
- Bioengineering Department, George Mason University, Fairfax, VA 22030, USA; (N.M.S.); (B.E.G.-G.); (H.D.)
| | - Blanca E. Gutiérrez-Guzmán
- Bioengineering Department, George Mason University, Fairfax, VA 22030, USA; (N.M.S.); (B.E.G.-G.); (H.D.)
| | - Holger Dannenberg
- Bioengineering Department, George Mason University, Fairfax, VA 22030, USA; (N.M.S.); (B.E.G.-G.); (H.D.)
- Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA 22030, USA
| | - Giorgio A. Ascoli
- Bioengineering Department, George Mason University, Fairfax, VA 22030, USA; (N.M.S.); (B.E.G.-G.); (H.D.)
- Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
9
|
Gong Y, Haeri M, Zhang X, Li Y, Liu A, Wu D, Zhang Q, Jazwinski SM, Zhou X, Wang X, Jiang L, Chen YP, Yan X, Swerdlow RH, Shen H, Deng HW. Spatial Dissection of the Distinct Cellular Responses to Normal Aging and Alzheimer's Disease in Human Prefrontal Cortex at Single-Nucleus Resolution. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.21.24306783. [PMID: 38826275 PMCID: PMC11142279 DOI: 10.1101/2024.05.21.24306783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Aging significantly elevates the risk for Alzheimer's disease (AD), contributing to the accumulation of AD pathologies, such as amyloid-β (Aβ), inflammation, and oxidative stress. The human prefrontal cortex (PFC) is highly vulnerable to the impacts of both aging and AD. Unveiling and understanding the molecular alterations in PFC associated with normal aging (NA) and AD is essential for elucidating the mechanisms of AD progression and developing novel therapeutics for this devastating disease. In this study, for the first time, we employed a cutting-edge spatial transcriptome platform, STOmics® SpaTial Enhanced Resolution Omics-sequencing (Stereo-seq), to generate the first comprehensive, subcellular resolution spatial transcriptome atlas of the human PFC from six AD cases at various neuropathological stages and six age, sex, and ethnicity matched controls. Our analyses revealed distinct transcriptional alterations across six neocortex layers, highlighted the AD-associated disruptions in laminar architecture, and identified changes in layer-to-layer interactions as AD progresses. Further, throughout the progression from NA to various stages of AD, we discovered specific genes that were significantly upregulated in neurons experiencing high stress and in nearby non-neuronal cells, compared to cells distant from the source of stress. Notably, the cell-cell interactions between the neurons under the high stress and adjacent glial cells that promote Aβ clearance and neuroprotection were diminished in AD in response to stressors compared to NA. Through cell-type specific gene co-expression analysis, we identified three modules in excitatory and inhibitory neurons associated with neuronal protection, protein dephosphorylation, and negative regulation of Aβ plaque formation. These modules negatively correlated with AD progression, indicating a reduced capacity for toxic substance clearance in AD subject samples. Moreover, we have discovered a novel transcription factor, ZNF460, that regulates all three modules, establishing it as a potential new therapeutic target for AD. Overall, utilizing the latest spatial transcriptome platform, our study developed the first transcriptome-wide atlas with subcellular resolution for assessing the molecular alterations in the human PFC due to AD. This atlas sheds light on the potential mechanisms underlying the progression from NA to AD.
Collapse
Affiliation(s)
- Yun Gong
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Mohammad Haeri
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, MO, 66160, USA
| | - Xiao Zhang
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Yisu Li
- Department of Cell and Molecular Biology, School of Science of Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Anqi Liu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Di Wu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Qilei Zhang
- School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410008, China
| | - S. Michal Jazwinski
- Tulane Center for Aging, Deming Department of Medicine, Tulane University School of Medicne, New Orleans, LA 70112, USA
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xiaoying Wang
- Clinical Neuroscience Research Center, Departments of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Lindong Jiang
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Yi-Ping Chen
- Department of Cell and Molecular Biology, School of Science of Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Xiaoxin Yan
- School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Russell H. Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, MO, 66160, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| |
Collapse
|
10
|
Sutton N, Gutiérrez-Guzmán B, Dannenberg H, Ascoli GA. A Continuous Attractor Model with Realistic Neural and Synaptic Properties Quantitatively Reproduces Grid Cell Physiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591748. [PMID: 38746202 PMCID: PMC11092518 DOI: 10.1101/2024.04.29.591748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Computational simulations with data-driven physiological detail can foster a deeper understanding of the neural mechanisms involved in cognition. Here, we utilize the wealth of cellular properties from Hippocampome.org to study neural mechanisms of spatial coding with a spiking continuous attractor network model of medial entorhinal cortex circuit activity. The primary goal was to investigate if adding such realistic constraints could produce firing patterns similar to those measured in real neurons. Biological characteristics included in the work are excitability, connectivity, and synaptic signaling of neuron types defined primarily by their axonal and dendritic morphologies. We investigate the spiking dynamics in specific neuron types and the synaptic activities between groups of neurons. Modeling the rodent hippocampal formation keeps the simulations to a computationally reasonable scale while also anchoring the parameters and results to experimental measurements. Our model generates grid cell activity that well matches the spacing, size, and firing rates of grid fields recorded in live behaving animals from both published datasets and new experiments performed for this study. Our simulations also recreate different scales of those properties, e.g., small and large, as found along the dorsoventral axis of the medial entorhinal cortex. Computational exploration of neuronal and synaptic model parameters reveals that a broad range of neural properties produce grid fields in the simulation. These results demonstrate that the continuous attractor network model of grid cells is compatible with a spiking neural network implementation sourcing data-driven biophysical and anatomical parameters from Hippocampome.org. The software is released as open source to enable broad community reuse and encourage novel applications.
Collapse
Affiliation(s)
- Nate Sutton
- Bioengineering Department, at George Mason University
| | | | - Holger Dannenberg
- Bioengineering Department, at George Mason University
- Interdisciplinary Program in Neuroscience at George Mason University
| | - Giorgio A. Ascoli
- Bioengineering Department, at George Mason University
- Interdisciplinary Program in Neuroscience at George Mason University
| |
Collapse
|
11
|
Shaheen H, Melnik R, Singh S. Data-driven Stochastic Model for Quantifying the Interplay Between Amyloid-beta and Calcium Levels in Alzheimer's Disease. Stat Anal Data Min 2024; 17:e11679. [PMID: 38646460 PMCID: PMC11031189 DOI: 10.1002/sam.11679] [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: 11/26/2023] [Accepted: 03/23/2024] [Indexed: 04/23/2024]
Abstract
The abnormal aggregation of extracellular amyloid-β ( A β ) in senile plaques resulting in calcium C a + 2 dyshomeostasis is one of the primary symptoms of Alzheimer's disease (AD). Significant research efforts have been devoted in the past to better understand the underlying molecular mechanisms driving A β deposition and C a + 2 dysregulation. Importantly, synaptic impairments, neuronal loss, and cognitive failure in AD patients are all related to the buildup of intraneuronal A β accumulation. Moreover, increasing evidence show a feed-forward loop between A β and C a + 2 levels, i.e. A β disrupts neuronal C a + 2 levels, which in turn affects the formation of A β . To better understand this interaction, we report a novel stochastic model where we analyze the positive feedback loop between A β and C a + 2 using ADNI data. A good therapeutic treatment plan for AD requires precise predictions. Stochastic models offer an appropriate framework for modelling AD since AD studies are observational in nature and involve regular patient visits. The etiology of AD may be described as a multi-state disease process using the approximate Bayesian computation method. So, utilizing ADNI data from 2-year visits for AD patients, we employ this method to investigate the interplay between A β and C a + 2 levels at various disease development phases. Incorporating the ADNI data in our physics-based Bayesian model, we discovered that a sufficiently large disruption in either A β metabolism or intracellular C a + 2 homeostasis causes the relative growth rate in both C a + 2 and A β , which corresponds to the development of AD. The imbalance of C a + 2 ions causes A β disorders by directly or indirectly affecting a variety of cellular and subcellular processes, and the altered homeostasis may worsen the abnormalities of C a + 2 ion transportation and deposition. This suggests that altering the C a + 2 balance or the balance between A β and C a + 2 by chelating them may be able to reduce disorders associated with AD and open up new research possibilities for AD therapy.
Collapse
Affiliation(s)
- Hina Shaheen
- Faculty of Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Roderick Melnik
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, Waterloo, ON N2L 3C5, Canada
| | - Sundeep Singh
- Faculty of Sustainable Design Engineering, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada
| | - The Alzheimer’s Disease Neuroimaging Initiative
- Data used in preparation of this article were generated by the Alzheimer’s Disease Metabolomics Consortium (ADMC). As such, the investigators within the ADMC provided data, but did not participate in the analysis or writing of this report. A complete listing of ADMC investigators can be found at: https://sites.duke.edu/adnimetab/team/
| |
Collapse
|
12
|
Zhang X, Chen C, Liu Y. Navigating the metabolic maze: anomalies in fatty acid and cholesterol processes in Alzheimer's astrocytes. Alzheimers Res Ther 2024; 16:63. [PMID: 38521950 PMCID: PMC10960454 DOI: 10.1186/s13195-024-01430-x] [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: 11/27/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, and its underlying mechanisms have been a subject of great interest. The mainstream theory of AD pathology suggests that the disease is primarily associated with tau protein and amyloid-beta (Aβ). However, an increasing body of research has revealed that abnormalities in lipid metabolism may be an important event throughout the pathophysiology of AD. Astrocytes, as important members of the lipid metabolism network in the brain, play a significant role in this event. The study of abnormal lipid metabolism in astrocytes provides a new perspective for understanding the pathogenesis of AD. This review focuses on the abnormal metabolism of fatty acids (FAs) and cholesterol in astrocytes in AD, and discusses it from three perspectives: lipid uptake, intracellular breakdown or synthesis metabolism, and efflux transport. We found that, despite the accumulation of their own fatty acids, astrocytes cannot efficiently uptake fatty acids from neurons, leading to fatty acid accumulation within neurons and resulting in lipotoxicity. In terms of cholesterol metabolism, astrocytes exhibit a decrease in endogenous synthesis due to the accumulation of exogenous cholesterol. Through a thorough investigation of these metabolic abnormalities, we can provide new insights for future therapeutic strategies by literature review to navigate this complex metabolic maze and bring hope to patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chuanying Chen
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Yi Liu
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
13
|
Wang P, Lynn A, Miskimen K, Song YE, Wisniewski T, Cohen M, Appleby BS, Safar JG, Haines JL. Genome-wide association studies identify novel loci in rapidly progressive Alzheimer's disease. Alzheimers Dement 2024; 20:2034-2046. [PMID: 38184787 PMCID: PMC10984493 DOI: 10.1002/alz.13655] [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: 06/14/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/08/2024]
Abstract
INTRODUCTION Recent data suggest that distinct prion-like amyloid beta and tau strains are associated with rapidly progressive Alzheimer's disease (rpAD). The role of genetic factors in rpAD is largely unknown. METHODS Previously known AD risk loci were examined in rpAD cases. Genome-wide association studies (GWAS) were performed to identify variants that influence rpAD. RESULTS We identified 115 pathology-confirmed rpAD cases and 193 clinical rpAD cases, 80% and 69% were of non-Hispanic European ancestry. Compared to the clinical cohort, pathology-confirmed rpAD had higher frequencies of apolipoprotein E (APOE) ε4 and rare missense variants in AD risk genes. A novel genome-wide significant locus (P < 5×10-8 ) was observed for clinical rpAD on chromosome 21 (rs2832546); 102 loci showed suggestive associations with pathology-confirmed rpAD (P < 1×10-5 ). DISCUSSION rpAD constitutes an extreme subtype of AD with distinct features. GWAS found previously known and novel loci associated with rpAD. Highlights Rapidly progressive Alzheimer's disease (rpAD) was defined with different criteria. Whole genome sequencing identified rare missense variants in rpAD. Novel variants were identified for clinical rpAD on chromosome 21.
Collapse
Affiliation(s)
- Ping Wang
- Department of Population and Quantitative Health SciencesSchool of Medicine, Case Western Reserve UniversityClevelandOhioUSA
| | - Audrey Lynn
- Department of Population and Quantitative Health SciencesSchool of Medicine, Case Western Reserve UniversityClevelandOhioUSA
- Cleveland Institute for Computational BiologyClevelandOhioUSA
| | - Kristy Miskimen
- Department of Population and Quantitative Health SciencesSchool of Medicine, Case Western Reserve UniversityClevelandOhioUSA
| | - Yeunjoo E. Song
- Department of Population and Quantitative Health SciencesSchool of Medicine, Case Western Reserve UniversityClevelandOhioUSA
| | - Thomas Wisniewski
- Departments of NeurologyPathology and PsychiatryCenter for Cognitive Neurology, NYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Mark Cohen
- Department of PathologyCase Western Reserve UniversityClevelandOhioUSA
- National Prion Disease Pathology Surveillance CenterCase Western Reserve UniversityClevelandOhioUSA
| | - Brian S. Appleby
- Department of PathologyCase Western Reserve UniversityClevelandOhioUSA
- National Prion Disease Pathology Surveillance CenterCase Western Reserve UniversityClevelandOhioUSA
- Department of NeurologyCase Western Reserve UniversityClevelandOhioUSA
- Department of PsychiatryCase Western Reserve UniversityClevelandOhioUSA
| | - Jiri G. Safar
- Department of PathologyCase Western Reserve UniversityClevelandOhioUSA
- Department of NeurologyCase Western Reserve UniversityClevelandOhioUSA
- Department of NeurosciencesCase Western Reserve UniversityClevelandOhioUSA
| | - Jonathan L. Haines
- Department of Population and Quantitative Health SciencesSchool of Medicine, Case Western Reserve UniversityClevelandOhioUSA
- Cleveland Institute for Computational BiologyClevelandOhioUSA
| |
Collapse
|
14
|
Malone TJ, Tien NW, Ma Y, Cui L, Lyu S, Wang G, Nguyen D, Zhang K, Myroshnychenko MV, Tyan J, Gordon JA, Kupferschmidt DA, Gu Y. A consistent map in the medial entorhinal cortex supports spatial memory. Nat Commun 2024; 15:1457. [PMID: 38368457 PMCID: PMC10874432 DOI: 10.1038/s41467-024-45853-4] [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: 09/20/2023] [Accepted: 02/05/2024] [Indexed: 02/19/2024] Open
Abstract
The medial entorhinal cortex (MEC) is hypothesized to function as a cognitive map for memory-guided navigation. How this map develops during learning and influences memory remains unclear. By imaging MEC calcium dynamics while mice successfully learned a novel virtual environment over ten days, we discovered that the dynamics gradually became more spatially consistent and then stabilized. Additionally, grid cells in the MEC not only exhibited improved spatial tuning consistency, but also maintained stable phase relationships, suggesting a network mechanism involving synaptic plasticity and rigid recurrent connectivity to shape grid cell activity during learning. Increased c-Fos expression in the MEC in novel environments further supports the induction of synaptic plasticity. Unsuccessful learning lacked these activity features, indicating that a consistent map is specific for effective spatial memory. Finally, optogenetically disrupting spatial consistency of the map impaired memory-guided navigation in a well-learned environment. Thus, we demonstrate that the establishment of a spatially consistent MEC map across learning both correlates with, and is necessary for, successful spatial memory.
Collapse
Affiliation(s)
- Taylor J Malone
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nai-Wen Tien
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Yan Ma
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lian Cui
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Shangru Lyu
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Garret Wang
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Duc Nguyen
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Center of Neural Science, New York University, New York, NY, USA
| | - Kai Zhang
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Maxym V Myroshnychenko
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jean Tyan
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Joshua A Gordon
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
- Office of the Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David A Kupferschmidt
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yi Gu
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
15
|
He Z, Zhang H, Li X, Shen L, Li N, Cheng S, Liu Q. Comparative proteomic analysis of cerebral cortex revealed neuroprotective mechanism of esculentoside A on Alzheimer's disease. Eur J Pharmacol 2024; 964:176226. [PMID: 38128868 DOI: 10.1016/j.ejphar.2023.176226] [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: 08/28/2023] [Revised: 11/09/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023]
Abstract
Esculentoside A (EsA), isolated from phytolacca esculenta, is a saponin showing neuroprotective effect in the mouse models of Alzheimer's disease (AD). To investigate its action target and underlying mechanism, this study used the proteomics technique of isobaric tags for relative and absolute quantification (iTRAQ) to analyze the differentially expressed proteins (DEPs) in the cerebral cortex of EsA-treated and untreated triple-transgenic 3 × Tg-AD model mice. Proteomic comparison revealed 250, 436, and 903 DEPs in three group pairs, i.e. AD/Wild-type (WT), AD+5 mg/kg EsA/AD, AD+10 mg/kg EsA/AD, respectively. Among them 28 DEPs were commonly shared by three group pairs, and 25 of them showed reversed expression levels in the diseased group under the treatment of both doses of EsA. Bioinformatics analysis revealed that these DEPs were mainly linked to metabolism, synapses, apoptosis, learning and memory. EsA treatment restored the expression of these proteins, including amyloid precursor protein (APP), cathepsin B (Cstb), 4-aminobutyrate aminotransferase (Abat), 3-phosphoinositide-dependent protein kinase-1 (PDK1), carnitine palmitoyltransferase1 (Cpt1) and synaptotagmin 17 (Syt17), thereby ameliorated the spatial learning and memory of AD mice. Collectively, this study reveals for the first time the profound effect of EsA on the cerebral cortex of AD mice, which might be a potential therapeutic agent for the treatment of AD.
Collapse
Affiliation(s)
- Zhijun He
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China; National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Huajie Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Xiaoqian Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Liming Shen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Nan Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, 518055, China
| | - Shuiyuan Cheng
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan, 430023, China.
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, 518055, China.
| |
Collapse
|
16
|
Nasb M, Tao W, Chen N. Alzheimer's Disease Puzzle: Delving into Pathogenesis Hypotheses. Aging Dis 2024; 15:43-73. [PMID: 37450931 PMCID: PMC10796101 DOI: 10.14336/ad.2023.0608] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disease characterized by both amnestic and non-amnestic clinical manifestations. It accounts for approximately 60-70% of all dementia cases worldwide. With the increasing number of AD patients, elucidating underlying mechanisms and developing corresponding interventional strategies are necessary. Hypotheses about AD such as amyloid cascade, Tau hyper-phosphorylation, neuroinflammation, oxidative stress, mitochondrial dysfunction, cholinergic, and vascular hypotheses are not mutually exclusive, and all of them play a certain role in the development of AD. The amyloid cascade hypothesis is currently the most widely studied; however, other hypotheses are also gaining support. This article summarizes the recent evidence regarding major pathological hypotheses of AD and their potential interplay, as well as the strengths and weaknesses of each hypothesis and their implications for the development of effective treatments. This could stimulate further studies and promote the development of more effective therapeutic strategies for AD.
Collapse
Affiliation(s)
| | | | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| |
Collapse
|
17
|
van Dyck CH, Mecca AP, O'Dell RS, Bartlett HH, Diepenbrock NG, Huang Y, Hamby ME, Grundman M, Catalano SM, Caggiano AO, Carson RE. A pilot study to evaluate the effect of CT1812 treatment on synaptic density and other biomarkers in Alzheimer's disease. Alzheimers Res Ther 2024; 16:20. [PMID: 38273408 PMCID: PMC10809445 DOI: 10.1186/s13195-024-01382-2] [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: 08/04/2023] [Accepted: 01/01/2024] [Indexed: 01/27/2024]
Abstract
BACKGROUND Effective, disease-modifying therapeutics for the treatment of Alzheimer's disease (AD) remain a large unmet need. Extensive evidence suggests that amyloid beta (Aβ) is central to AD pathophysiology, and Aβ oligomers are among the most toxic forms of Aβ. CT1812 is a novel brain penetrant sigma-2 receptor ligand that interferes with the binding of Aβ oligomers to neurons. Preclinical studies of CT1812 have demonstrated its ability to displace Aβ oligomers from neurons, restore synapses in cell cultures, and improve cognitive measures in mouse models of AD. CT1812 was found to be generally safe and well tolerated in a placebo-controlled phase 1 clinical trial in healthy volunteers and phase 1a/2 clinical trials in patients with mild to moderate dementia due to AD. The unique objective of this study was to incorporate synaptic positron emission tomography (PET) imaging as an outcome measure for CT1812 in AD patients. METHODS The present phase 1/2 study was a randomized, double-blind, placebo-controlled, parallel-group trial conducted in 23 participants with mild to moderate dementia due to AD to primarily evaluate the safety of CT1812 and secondarily its pharmacodynamic effects. Participants received either placebo or 100 mg or 300 mg per day of oral CT1812 for 24 weeks. Pharmacodynamic effects were assessed using the exploratory efficacy endpoints synaptic vesicle glycoprotein 2A (SV2A) PET, fluorodeoxyglucose (FDG) PET, volumetric MRI, cognitive clinical measures, as well as cerebrospinal fluid (CSF) biomarkers of AD pathology and synaptic degeneration. RESULTS No treatment differences relative to placebo were observed in the change from baseline at 24 weeks in either SV2A or FDG PET signal, the cognitive clinical rating scales, or in CSF biomarkers. Composite region volumetric MRI revealed a trend towards tissue preservation in participants treated with either dose of CT1812, and nominally significant differences with both doses of CT1812 compared to placebo were found in the pericentral, prefrontal, and hippocampal cortices. CT1812 was safe and well tolerated. CONCLUSIONS The safety findings of this 24-week study and the observed changes on volumetric MRI with CT1812 support its further clinical development. TRIAL REGISTRATION The clinical trial described in this manuscript is registered at clinicaltrials.gov (NCT03493282).
Collapse
Affiliation(s)
- Christopher H van Dyck
- Alzheimer's Disease Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| | - Adam P Mecca
- Alzheimer's Disease Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Ryan S O'Dell
- Alzheimer's Disease Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Hugh H Bartlett
- Alzheimer's Disease Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Nina G Diepenbrock
- Alzheimer's Disease Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Mary E Hamby
- Cognition Therapeutics Inc., Pittsburgh, PA, USA
| | - Michael Grundman
- Global R&D Partners, LLC, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego, USA
| | | | | | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| |
Collapse
|
18
|
Zhao F, Ma X, Yao B, Chen L. scaDA: A Novel Statistical Method for Differential Analysis of Single-Cell Chromatin Accessibility Sequencing Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.21.576570. [PMID: 38328112 PMCID: PMC10849518 DOI: 10.1101/2024.01.21.576570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Single-cell ATAC-seq sequencing data (scATAC-seq) has been widely used to investigate chromatin accessibility on the single-cell level. One important application of scATAC-seq data analysis is differential chromatin accessibility analysis. However, the data characteristics of scATAC-seq such as excessive zeros and large variability of chromatin accessibility across cells impose a unique challenge for DA analysis. Existing statistical methods focus on detecting the mean difference of the chromatin accessible regions while overlooking the distribution difference. Motivated by real data exploration that distribution difference exists among cell types, we introduce a novel composite statistical test named "scaDA", which is based on zero-inflated negative binomial model (ZINB), for performing differential distribution analysis of chromatin accessibility by jointly testing the abundance, prevalence and dispersion simultaneously. Benefiting from both dispersion shrinkage and iterative refinement of mean and prevalence parameter estimates, scaDA demonstrates its superiority to both ZINB-based likelihood ratio tests and published methods by achieving the highest power and best FDR control in a comprehensive simulation study. In addition to demonstrating the highest power in three real sc-multiome data analyses, scaDA successfully identifies differentially accessible regions in microglia from sc-multiome data for an Alzheimer's disease (AD) study, regions which are most enriched in GO terms related to neurogenesis, the clinical phenotype of AD, and SNPs identified in AD-associated GWAS.
Collapse
Affiliation(s)
- Fengdi Zhao
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Xin Ma
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Bing Yao
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Li Chen
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
19
|
Bae H, Kang MJ, Ha SW, Jeong DE, Lee K, Lim S, Min JY, Min KB. Association of plasma amyloid-β oligomerization with theta/beta ratio in older adults. Front Aging Neurosci 2023; 15:1291881. [PMID: 38106526 PMCID: PMC10722169 DOI: 10.3389/fnagi.2023.1291881] [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: 09/10/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023] Open
Abstract
Background Oligomeric Aβ (OAβ) is a promising candidate marker for Alzheimer's disease (AD) diagnosis. Electroencephalography (EEG) is a potential tool for early detection of AD. Still, whether EEG power ratios, particularly the theta/alpha ratio (TAR) and theta/beta ratio (TBR), reflect Aβ burden-a primary mechanism underlying cognitive impairment and AD. This study investigated the association of TAR and TBR with amyloid burden in older adults based on MDS-OAβ levels. Methods 529 individuals (aged ≥60 years) were recruited. All participants underwent EEG (MINDD SCAN, Ybrain Inc., South Korea) and AlzOn™ test (PeopleBio Inc., Gyeonggi-do, Republic of Korea) for quantifying MDS-OAβ values in the plasma. EEG variables were log-transformed to normalize the data distribution. Using the MDS-OAβ cutoff value (0.78 ng/mL), all participants were classified into two groups: high MDS-OAβ and low MDS-OAβ. Results Participants with high MDS-OAβ levels had significantly higher TARs and TBRs than those with low MDS-OAβ levels. The log-transformed TBRs in the central lobe (β = 0.161, p = 0.0026), frontal lobe (β = 0.145, p = 0.0044), parietal lobe (β = 0.166, p = 0.0028), occipital lobe (β = 0.158, p = 0.0058), and temporal lobe (beta = 0.162, p = 0.0042) were significantly and positively associated with increases in MDS-OAβ levels. After adjusting for the Bonferroni correction, the TBRs in all lobe regions, except the occipital lobe, were significantly associated with increased MDS-OAβ levels. Conclusion We found a significant association of MDS-OAβ with TBR in older adults. This finding indicates that an increase in amyloid burden may be associated with an increase in the low-frequency band and a decrease in the relatively high-frequency band.
Collapse
Affiliation(s)
- Heewon Bae
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Min Ju Kang
- Department of Neurology, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Sang-Won Ha
- Department of Neurology, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Da-Eun Jeong
- Department of Neurology, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Kiwon Lee
- Ybrain Research Institute, Seongnam-si, Republic of Korea
| | - Seungui Lim
- Ybrain Research Institute, Seongnam-si, Republic of Korea
| | - Jin-Young Min
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Kyoung-Bok Min
- Department of Preventive Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Medical Research Center, Institute of Health Policy and Management, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
20
|
Dalva MB. Brain cancer thrives by hijacking mechanisms to boost synapse strength. Nature 2023; 623:260-262. [PMID: 37932559 DOI: 10.1038/d41586-023-03306-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
|
21
|
Xu M, Liu Q, Bi R, Li Y, Li H, Kang WB, Yan Z, Zheng Q, Sun C, Ye M, Xiang BL, Luo XJ, Li M, Zhang DF, Yao YG. Coexistence of Multiple Functional Variants and Genes Underlies Genetic Risk Locus 11p11.2 of Alzheimer's Disease. Biol Psychiatry 2023; 94:743-759. [PMID: 37290560 DOI: 10.1016/j.biopsych.2023.05.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND Genome-wide association studies have identified dozens of genetic risk loci for Alzheimer's disease (AD), yet the underlying causal variants and biological mechanisms remain elusive, especially for loci with complex linkage disequilibrium and regulation. METHODS To fully untangle the causal signal at a single locus, we performed a functional genomic study of 11p11.2 (the CELF1/SPI1 locus). Genome-wide association study signals at 11p11.2 were integrated with datasets of histone modification, open chromatin, and transcription factor binding to distill potentially functional variants (fVars). Their allelic regulatory activities were confirmed by allele imbalance, reporter assays, and base editing. Expressional quantitative trait loci and chromatin interaction data were incorporated to assign target genes to fVars. The relevance of these genes to AD was assessed by convergent functional genomics using bulk brain and single-cell transcriptomic, epigenomic, and proteomic datasets of patients with AD and control individuals, followed by cellular assays. RESULTS We found that 24 potential fVars, rather than a single variant, were responsible for the risk of 11p11.2. These fVars modulated transcription factor binding and regulated multiple genes by long-range chromatin interactions. Besides SPI1, convergent evidence indicated that 6 target genes (MTCH2, ACP2, NDUFS3, PSMC3, C1QTNF4, and MADD) of fVars were likely to be involved in AD development. Disruption of each gene led to cellular amyloid-β and phosphorylated tau changes, supporting the existence of multiple likely causal genes at 11p11.2. CONCLUSIONS Multiple variants and genes at 11p11.2 may contribute to AD risk. This finding provides new insights into the mechanistic and therapeutic challenges of AD.
Collapse
Affiliation(s)
- Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Qianjin Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Rui Bi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China; National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Yu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Hongli Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Wei-Bo Kang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Zhongjiang Yan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Quanzhen Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Chunli Sun
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Maosen Ye
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Bo-Lin Xiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Deng-Feng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China; National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China; National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
22
|
Lu K, Li C, Liu J, Wang J, Li Y, He B, Li J, Zhang X, Wei M, Tian Y, Zhang R, Zhang C, Zhang Y. Impairments in endogenous AMPA receptor dynamics correlates with learning deficits in Alzheimer's disease model mice. Proc Natl Acad Sci U S A 2023; 120:e2303878120. [PMID: 37748061 PMCID: PMC10556575 DOI: 10.1073/pnas.2303878120] [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: 03/27/2023] [Accepted: 08/29/2023] [Indexed: 09/27/2023] Open
Abstract
AMPA receptors (AMPARs) play a critical role in synaptic plasticity and learning and memory, and dysfunction or dysregulation of AMPARs could lead to various neurological and psychiatric disorders, such as Alzheimer's disease (AD). However, the dynamics and/or longitudinal changes of AMPARs in vivo during AD pathogenesis remain elusive. Here, employing 5xFAD SEP-GluA1 KI mice, we investigated endogenous AMPA receptor dynamics in a whisker deflection-associated Go/No-go learning paradigm. We found a significant increase in synaptosomal AMPA receptor subunits GluA1 in WT mice after learning, while no such changes were detected in 7-mo-old 5xFAD mice. Daily training led to an increase in endogenous spine surface GluA1 in Control mice, while this increase was absent in 5xFAD-KI mice which correlates with its learning defects in Go/No-go paradigm. Furthermore, we demonstrated that the onset of abnormal AMPAR dynamics corresponds temporally with microglia and astrocyte overactivation. Our results have shown that impairments in endogenous AMPA receptor dynamics play an important role in learning deficits in 5xFAD mice and AD pathogenesis.
Collapse
Affiliation(s)
- Kongjie Lu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Chenyang Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Jiao Liu
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing100083, China
| | - Jinpeng Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Yongfeng Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Bin He
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Junzhao Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Xiaochen Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin300072, China
| | - Mengping Wei
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing100069, China
| | - Yonglu Tian
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
- School of Psychological and Cognitive Sciences, Peking University, Beijing100871, China
| | - Rong Zhang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing100069, China
| | - Yong Zhang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of the People’s Republic of China, Beijing100083, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing100871, China
| |
Collapse
|
23
|
Malone TJ, Tien NW, Ma Y, Cui L, Lyu S, Wang G, Nguyen D, Zhang K, Myroshnychenko MV, Tyan J, Gordon JA, Kupferschmidt DA, Gu Y. A consistent map in the medial entorhinal cortex supports spatial memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.560254. [PMID: 37986767 PMCID: PMC10659391 DOI: 10.1101/2023.09.30.560254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The medial entorhinal cortex (MEC) is hypothesized to function as a cognitive map for memory-guided navigation. How this map develops during learning and influences memory remains unclear. By imaging MEC calcium dynamics while mice successfully learned a novel virtual environment over ten days, we discovered that the dynamics gradually became more spatially consistent and then stabilized. Additionally, grid cells in the MEC not only exhibited improved spatial tuning consistency, but also maintained stable phase relationships, suggesting a network mechanism involving synaptic plasticity and rigid recurrent connectivity to shape grid cell activity during learning. Increased c-Fos expression in the MEC in novel environments further supports the induction of synaptic plasticity. Unsuccessful learning lacked these activity features, indicating that a consistent map is specific for effective spatial memory. Finally, optogenetically disrupting spatial consistency of the map impaired memory-guided navigation in a well-learned environment. Thus, we demonstrate that the establishment of a spatially consistent MEC map across learning both correlates with, and is necessary for, successful spatial memory.
Collapse
Affiliation(s)
- Taylor J. Malone
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- These authors contributed equally to this work
| | - Nai-Wen Tien
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Current address: Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- These authors contributed equally to this work
| | - Yan Ma
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- These authors contributed equally to this work
| | - Lian Cui
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shangru Lyu
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Garret Wang
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Duc Nguyen
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Current address: Center of Neural Science, New York University, New York, NY, USA
| | - Kai Zhang
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Maxym V. Myroshnychenko
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jean Tyan
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joshua A. Gordon
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
- Office of the Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - David A. Kupferschmidt
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yi Gu
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
24
|
Rejdak K, Sienkiewicz-Jarosz H, Bienkowski P, Alvarez A. Modulation of neurotrophic factors in the treatment of dementia, stroke and TBI: Effects of Cerebrolysin. Med Res Rev 2023; 43:1668-1700. [PMID: 37052231 DOI: 10.1002/med.21960] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023]
Abstract
Neurotrophic factors (NTFs) are involved in the pathophysiology of neurological disorders such as dementia, stroke and traumatic brain injury (TBI), and constitute molecular targets of high interest for the therapy of these pathologies. In this review we provide an overview of current knowledge of the definition, discovery and mode of action of five NTFs, nerve growth factor, insulin-like growth factor 1, brain derived NTF, vascular endothelial growth factor and tumor necrosis factor alpha; as well as on their contribution to brain pathology and potential therapeutic use in dementia, stroke and TBI. Within the concept of NTFs in the treatment of these pathologies, we also review the neuropeptide preparation Cerebrolysin, which has been shown to resemble the activities of NTFs and to modulate the expression level of endogenous NTFs. Cerebrolysin has demonstrated beneficial treatment capabilities in vitro and in clinical studies, which are discussed within the context of the biochemistry of NTFs. The review focuses on the interactions of different NTFs, rather than addressing a single NTF, by outlining their signaling network and by reviewing their effect on clinical outcome in prevalent brain pathologies. The effects of the interactions of these NTFs and Cerebrolysin on neuroplasticity, neurogenesis, angiogenesis and inflammation, and their relevance for the treatment of dementia, stroke and TBI are summarized.
Collapse
Affiliation(s)
- Konrad Rejdak
- Department of Neurology, Medical University of Lublin, Lublin, Poland
| | | | | | - Anton Alvarez
- Medinova Institute of Neurosciences, Clinica RehaSalud, Coruña, Spain
| |
Collapse
|
25
|
Janković T, Pilipović K. Single Versus Repetitive Traumatic Brain Injury: Current Knowledge on the Chronic Outcomes, Neuropathology and the Role of TDP-43 Proteinopathy. Exp Neurobiol 2023; 32:195-215. [PMID: 37749924 PMCID: PMC10569144 DOI: 10.5607/en23008] [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: 02/16/2023] [Revised: 07/18/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the most important causes of death and disability in adults and thus an important public health problem. Following TBI, secondary pathophysiological processes develop over time and condition the development of different neurodegenerative entities. Previous studies suggest that neurobehavioral changes occurring after a single TBI are the basis for the development of Alzheimer's disease, while repetitive TBI is considered to be a contributing factor for chronic traumatic encephalopathy development. However, pathophysiological processes that determine the evolvement of a particular chronic entity are still unclear. Human post-mortem studies have found combinations of amyloid, tau, Lewi bodies, and TAR DNA-binding protein 43 (TDP-43) pathologies after both single and repetitive TBI. This review focuses on the pathological changes of TDP-43 after single and repetitive brain traumas. Numerous studies have shown that TDP-43 proteinopathy noticeably occurs after repetitive head trauma. A relatively small number of available preclinical research on single brain injury are not in complete agreement with the results from the human samples, which makes it difficult to draw specific conclusions. Also, as TBI is considered a heterogeneous type of injury, different experimental trauma models and injury intensities may cause differences in the cascade of secondary injury, which should be considered in future studies. Experimental and post-mortem studies of TDP-43 pathobiology should be carried out, preferably in the same laboratories, to determine its involvement in the development of neurodegenerative conditions after one and repetitive TBI, especially in the context of the development of new therapeutic options.
Collapse
Affiliation(s)
- Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Rijeka 51000, Croatia
| |
Collapse
|
26
|
Zhang NK, Zhang SK, Zhang LI, Tao HW, Zhang GW. Sensory processing deficits and related cortical pathological changes in Alzheimer's disease. Front Aging Neurosci 2023; 15:1213379. [PMID: 37649717 PMCID: PMC10464619 DOI: 10.3389/fnagi.2023.1213379] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/24/2023] [Indexed: 09/01/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder primarily affecting cognitive functions. However, sensory deficits in AD start to draw attention due to their high prevalence and early onsets which suggest that they could potentially serve as diagnostic biomarkers and even contribute to the disease progression. This literature review examines the sensory deficits and cortical pathological changes observed in visual, auditory, olfactory, and somatosensory systems in AD patients, as well as in various AD animal models. Sensory deficits may emerge at the early stages of AD, or even precede the cognitive decline, which is accompanied by cortical pathological changes including amyloid-beta deposition, tauopathy, gliosis, and alterations in neuronal excitability, synaptic inputs, and functional plasticity. Notably, these changes are more pronounced in sensory association areas and superficial cortical layers, which may explain the relative preservation of basic sensory functions but early display of deficits of higher sensory functions. We propose that sensory impairment and the progression of AD may establish a cyclical relationship that mutually perpetuates each condition. This review highlights the significance of sensory deficits with or without cortical pathological changes in AD and emphasizes the need for further research to develop reliable early detection and intervention through sensory systems.
Collapse
Affiliation(s)
- Nicole K. Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Selena K. Zhang
- Biomedical Engineering Program, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Li I. Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Physiology & Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Huizhong W. Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Physiology & Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
27
|
Wang Y, Ye X, Song B, Yan Y, Ma W, Shi J. Features of event-related potentials during retrieval of episodic memory in patients with mild cognitive impairment due to Alzheimer's disease. Front Neurosci 2023; 17:1185228. [PMID: 37469837 PMCID: PMC10352679 DOI: 10.3389/fnins.2023.1185228] [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: 03/13/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023] Open
Abstract
Objective To provide a rigorous comparison between patients with mild cognitive impairment due to Alzheimer's disease (MCI-AD) and healthy elderly, as well as to assess the value of electroencephalography (EEG) in terms of early diagnosis, we conducted a neutral image recognition memory task involving individuals with positive biomarkers including β amyloid deposition, pathologic tau or neurodegeneration. Methods The task involving study and test blocks was designed to evaluate participants' recognition memory. Electroencephalogram was recorded synchronously to elicit event-related potentials in patients with MCI-AD and healthy control subjects. We further analyzed differences between groups or conditions in terms of behavioral performance, time domain, and time-frequency domain. Results The MCI-AD cohort showed a slower response time to old/new images and had low accuracy regarding behavioral performance. The amplitude of the late positive complex for the old/new effects was significantly suppressed in the MCI-AD cohort when compared with that in the HC cohort. The amplitude of the late old/new effects was correlated with the Auditory Verbal Learning Test recognition score in all participants. The time-frequency domain analysis revealed that correct recognition of old items elicited a decrease in beta power, mainly limited to the HC cohort. Moreover, the combination of behavioral (processing speed and accuracy) and electrophysiological (average amplitude and relative power of delta band) measures contributes to classifying patients with MCI-AD from healthy elderly people. Conclusion Changes of old/new effects, accuracy and response time are sensitive to the impairment of recognition memory in patients with MCI-AD and have moderate value in predicting the incipient stage of AD.
Collapse
|
28
|
Na H, Yang JB, Zhang Z, Gan Q, Tian H, Rajab IM, Potempa LA, Tao Q, Qiu WQ. Peripheral apolipoprotein E proteins and their binding to LRP1 antagonize Alzheimer's disease pathogenesis in the brain during peripheral chronic inflammation. Neurobiol Aging 2023; 127:54-69. [PMID: 37060729 PMCID: PMC10198819 DOI: 10.1016/j.neurobiolaging.2023.02.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 12/15/2022] [Accepted: 02/28/2023] [Indexed: 03/09/2023]
Abstract
C-reactive protein (CRP) impacts apolipoprotein E4 (ApoE4) allele to increase Alzheimer's disease (AD) risk. However, it is unclear how the ApoE protein and its binding to LRP1 are involved. We found that ApoE2 carriers had the highest but ApoE4 carriers had the lowest concentrations of blood ApoE in both humans and mice; blood ApoE concentration was negatively associated with AD risk. Elevation of peripheral monomeric CRP (mCRP) reduced the expression of ApoE in ApoE2 mice, while it decreased ApoE-LRP1 binding in the brains of ApoE4 mice that was characterized by Proximity Ligation Assay. Both serum ApoE and brain ApoE-LRP1 binding were positively associated with the expression of pericytes that disappeared after mCRP treatment, and negatively associated with brain tauopathy and neuroinflammation in the presence of mCRP. In ApoE-/- mice, mCRP reduced the brain expression levels of synaptophysin and PSD95 and the positive relationship between ApoE-LRP1 binding and synaptophysin or PSD95 expression disappeared. Our study suggests that blood ApoE protects against AD pathogenesis by binding to LRP1 during peripheral chronic inflammation.
Collapse
Affiliation(s)
- Hana Na
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Jack B Yang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Zhengrong Zhang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Qini Gan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Hua Tian
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA; Department of Pharmacology, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | | | | | - Qiushan Tao
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA; Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA; Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
29
|
Ahmed A, Zeng G, Azhar M, Wang F, Wang J, Fan B, Liu X, Jiang D, Wang Q. Combination of Shengmai San and Radix puerariae ameliorates depression-like symptoms in diabetic rats at the nexus of PI3K/BDNF/SYN protein expression. Animal Model Exp Med 2023; 6:211-220. [PMID: 37317044 PMCID: PMC10272924 DOI: 10.1002/ame2.12333] [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: 12/09/2022] [Accepted: 04/03/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Hyperglycemia is a characteristic feature of diabetes that often results in neuropsychological complications such as depression. Diabetic individuals are more vulnerable to experience depression compared to the normal population. Thus, novel treatment approaches are required to reduce depressive symptoms among diabetic individuals. Traditional Chinese medicines (TCMs) such as Shengmai San (SMS) and Radix puerariae (R) are usually widely used to treat ailments such as neurological complications since ancient time. METHODS In this study, SMS was combined with R to prepare an R-SMS formulation and screened for their antidepressant activity in diabetic rats. The antidepressant potential of the prepared combination was evaluated behaviorally using open field test, novelty-induced hypophagia, and forced swim test in diabetic rats with biochemical and protein expression (PI3K, BDNF [brain-derived neurotrophic factor], and SYN [presynaptic vesicle protein]) analysis. RESULTS Diabetic rats (streptozotocin, 45 mg/kg) showed elevated fasting blood glucose (FBG) >12 mM with depressive symptoms throughout the study. Treatment with R-SMS (0.5, 1.5, and 4.5 g/kg) significantly reverted depressive symptoms in diabetic rats as evinced by significantly (p < 0.05) reduced immobility time with an increased tendency to eat food in a novel environment. Treatment with R-SMS also significantly increased the protein expression of PI3K, BDNF, and SYN protein, which play a crucial role in depression. CONCLUSION This study showed that R-SMS formulation antagonized depressive symptoms in diabetic rats; thus, this formulation might be studied further to develop as an antidepressant.
Collapse
Affiliation(s)
- Ayaz Ahmed
- Institute of Food Science and TechnologyChinese Academy of Agricultural SciencesBeijingChina
- Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs & Hunan Provincial Research Center for Safety Evaluation of DrugsChangshaChina
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Guirong Zeng
- Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs & Hunan Provincial Research Center for Safety Evaluation of DrugsChangshaChina
- Institute of Drug Discovery TechnologyNingbo UniversityNingboChina
- Research Center for Pharmacodynamic, Material Basis and Mechanism of ActionCollege of Pharmacy, Guizhou University of Traditional Chinese MedicineGuiyangChina
| | - Mudassar Azhar
- Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs & Hunan Provincial Research Center for Safety Evaluation of DrugsChangshaChina
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Fengzhong Wang
- Institute of Food Science and TechnologyChinese Academy of Agricultural SciencesBeijingChina
| | - Jingru Wang
- Research Center for Pharmacodynamic, Material Basis and Mechanism of ActionCollege of Pharmacy, Guizhou University of Traditional Chinese MedicineGuiyangChina
| | - Bei Fan
- Institute of Food Science and TechnologyChinese Academy of Agricultural SciencesBeijingChina
| | - Xinmin Liu
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
- Institute of Drug Discovery TechnologyNingbo UniversityNingboChina
| | - Dejiang Jiang
- Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs & Hunan Provincial Research Center for Safety Evaluation of DrugsChangshaChina
| | - Qiong Wang
- Institute of Food Science and TechnologyChinese Academy of Agricultural SciencesBeijingChina
- Sino‐Portugal TCM International Cooperation CenterThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
30
|
García-Rodríguez C, Mujica P, Illanes-González J, López A, Vargas C, Sáez JC, González-Jamett A, Ardiles ÁO. Probenecid, an Old Drug with Potential New Uses for Central Nervous System Disorders and Neuroinflammation. Biomedicines 2023; 11:1516. [PMID: 37371611 DOI: 10.3390/biomedicines11061516] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
Probenecid is an old uricosuric agent used in clinics to treat gout and reduce the renal excretion of antibiotics. In recent years, probenecid has gained attention due to its ability to interact with membrane proteins such as TRPV2 channels, organic anion transporters, and pannexin 1 hemichannels, which suggests new potential therapeutic utilities in medicine. Some current functions of probenecid include their use as an adjuvant to increase the bioavailability of several drugs in the Central Nervous System (CNS). Numerous studies also suggest that this drug has important neuroprotective, antiepileptic, and anti-inflammatory properties, as evidenced by their effect against neurological and neurodegenerative diseases. In these studies, the use of probenecid as a Panx1 hemichannel blocker to reduce neuroinflammation is highlighted since neuroinflammation is a major trigger for diverse CNS alterations. Although the clinical use of probenecid has declined over the years, advances in its use in preclinical research indicate that it may be useful to improve conventional therapies in the psychiatric field where the drugs used have a low bioavailability, either because of a deficient passage through the blood-brain barrier or a high efflux from the CNS or also a high urinary clearance. This review summarizes the history, pharmacological properties, and recent research uses of probenecid and discusses its future projections as a potential pharmacological strategy to intervene in neurodegeneration as an outcome of neuroinflammation.
Collapse
Affiliation(s)
- Claudia García-Rodríguez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Paula Mujica
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Javiera Illanes-González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Araceli López
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Camilo Vargas
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Juan C Sáez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Arlek González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Álvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| |
Collapse
|
31
|
Liu M, Zhu L, Guo YJ, Zhang SS, Jiang L, Zhang Y, Chao FL, Tang Y. The effects of voluntary running exercise on the astrocytes of the medial prefrontal cortex in APP/PS1 mice. J Comp Neurol 2023. [PMID: 37146123 DOI: 10.1002/cne.25485] [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: 11/27/2022] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 05/07/2023]
Abstract
Pathological changes in the medial prefrontal cortex (mPFC) and astrocytes are closely associated with Alzheimer's disease (AD). Voluntary running has been found to effectively delay AD. However, the effects of voluntary running on mPFC astrocytes in AD are unclear. A total of 40 10-month-old male amyloid precursor protein/presenilin 1 (APP/PS1) mice and 40 wild-type (WT) mice were randomly divided into control and running groups, and the running groups underwent voluntary running for 3 months. Mouse cognition was assessed by the novel object recognition (NOR), Morris water maze (MWM), and Y maze tests. The effects of voluntary running on mPFC astrocytes were investigated using immunohistochemistry, immunofluorescence, western blotting, and stereology. APP/PS1 mice performed significantly worse than WT mice in the NOR, MWM, and Y maze tests, and voluntary running improved the performance of APP/PS1 mice in these tests. The total number of mPFC astrocytes was increased, cell bodies were enlarged, and protrusion number and length were increased in AD mice compared with WT mice, but there was no difference in component 3 (C3) levels in the mPFC (total mPFC level); however, C3 and S100B levels in astrocytes were increased in AD mice. Voluntary running reduced the total number of astrocytes and S100B levels in astrocytes and increased the density of PSD95+ puncta in direct contact with astrocyte protrusions in the APP/PS1 mouse mPFC. Three months of voluntary running inhibited astrocyte hyperplasia and S100B expression in astrocytes, increased the density of synapses in contact with astrocytes, and improved cognitive function in APP/PS1 mice.
Collapse
Affiliation(s)
- Mei Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Lin Zhu
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Yi-Jing Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Shan-Shan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Army Medical University, Chongqing, P. R. China
| | - Lin Jiang
- Laboratory Teaching & Management Center, Chongqing Medical University, Chongqing, P. R. China
| | - Yi Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Feng-Lei Chao
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Yong Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
32
|
Feller B, Fallon A, Luo W, Nguyen PT, Shlaifer I, Lee AK, Chofflet N, Yi N, Khaled H, Karkout S, Bourgault S, Durcan TM, Takahashi H. α-Synuclein Preformed Fibrils Bind to β-Neurexins and Impair β-Neurexin-Mediated Presynaptic Organization. Cells 2023; 12:cells12071083. [PMID: 37048156 PMCID: PMC10093570 DOI: 10.3390/cells12071083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Synucleinopathies form a group of neurodegenerative diseases defined by the misfolding and aggregation of α-synuclein (α-syn). Abnormal accumulation and spreading of α-syn aggregates lead to synapse dysfunction and neuronal cell death. Yet, little is known about the synaptic mechanisms underlying the α-syn pathology. Here we identified β-isoforms of neurexins (β-NRXs) as presynaptic organizing proteins that interact with α-syn preformed fibrils (α-syn PFFs), toxic α-syn aggregates, but not α-syn monomers. Our cell surface protein binding assays and surface plasmon resonance assays reveal that α-syn PFFs bind directly to β-NRXs through their N-terminal histidine-rich domain (HRD) at the nanomolar range (KD: ~500 nM monomer equivalent). Furthermore, our artificial synapse formation assays show that α-syn PFFs diminish excitatory and inhibitory presynaptic organization induced by a specific isoform of neuroligin 1 that binds only β-NRXs, but not α-isoforms of neurexins. Thus, our data suggest that α-syn PFFs interact with β-NRXs to inhibit β-NRX-mediated presynaptic organization, providing novel molecular insight into how α-syn PFFs induce synaptic pathology in synucleinopathies such as Parkinson’s disease and dementia with Lewy bodies.
Collapse
Affiliation(s)
- Benjamin Feller
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Aurélie Fallon
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Wen Luo
- The Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Phuong Trang Nguyen
- Department of Chemistry, Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada
| | - Irina Shlaifer
- The Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Alfred Kihoon Lee
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B2, Canada
| | - Nicolas Chofflet
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B2, Canada
| | - Nayoung Yi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Husam Khaled
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Samer Karkout
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
| | - Steve Bourgault
- Department of Chemistry, Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada
| | - Thomas M. Durcan
- The Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B2, Canada
| | - Hideto Takahashi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B2, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
33
|
Ye C, Behnke JA, Hardin KR, Zheng JQ. Drosophila melanogaster as a model to study age and sex differences in brain injury and neurodegeneration after mild head trauma. Front Neurosci 2023; 17:1150694. [PMID: 37077318 PMCID: PMC10106652 DOI: 10.3389/fnins.2023.1150694] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/09/2023] [Indexed: 04/05/2023] Open
Abstract
Repetitive physical insults to the head, including those that elicit mild traumatic brain injury (mTBI), are a known risk factor for a variety of neurodegenerative conditions including Alzheimer's disease (AD), Parkinson's disease (PD), and chronic traumatic encephalopathy (CTE). Although most individuals who sustain mTBI typically achieve a seemingly full recovery within a few weeks, a subset experience delayed-onset symptoms later in life. As most mTBI research has focused on the acute phase of injury, there is an incomplete understanding of mechanisms related to the late-life emergence of neurodegeneration after early exposure to mild head trauma. The recent adoption of Drosophila-based brain injury models provides several unique advantages over existing preclinical animal models, including a tractable framework amenable to high-throughput assays and short relative lifespan conducive to lifelong mechanistic investigation. The use of flies also provides an opportunity to investigate important risk factors associated with neurodegenerative conditions, specifically age and sex. In this review, we survey current literature that examines age and sex as contributing factors to head trauma-mediated neurodegeneration in humans and preclinical models, including mammalian and Drosophila models. We discuss similarities and disparities between human and fly in aging, sex differences, and pathophysiology. Finally, we highlight Drosophila as an effective tool for investigating mechanisms underlying head trauma-induced neurodegeneration and for identifying therapeutic targets for treatment and recovery.
Collapse
Affiliation(s)
- Changtian Ye
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Joseph A. Behnke
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Katherine R. Hardin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - James Q. Zheng
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
34
|
Lee AK, Yi N, Khaled H, Feller B, Takahashi H. SorCS1 inhibits amyloid-β binding to neurexin and rescues amyloid-β-induced synaptic pathology. Life Sci Alliance 2023; 6:e202201681. [PMID: 36697254 PMCID: PMC9880023 DOI: 10.26508/lsa.202201681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Amyloid-β oligomers (AβOs), toxic peptide aggregates found in Alzheimer's disease, cause synapse pathology. AβOs interact with neurexins (NRXs), key synaptic organizers, and this interaction dampens normal trafficking and function of NRXs. Axonal trafficking of NRX is in part regulated by its interaction with SorCS1, a protein sorting receptor, but the impact of SorCS1 regulation of NRXs in Aβ pathology was previously unstudied. Here, we show competition between the SorCS1 ectodomain and AβOs for β-NRX binding and rescue effects of the SorCS1b isoform on AβO-induced synaptic pathology. Like AβOs, the SorCS1 ectodomain binds to NRX1β through the histidine-rich domain of NRX1β, and the SorCS1 ectodomain and AβOs compete for NRX1β binding. In cultured hippocampal neurons, SorCS1b colocalizes with NRX1β on the axon surface, and axonal expression of SorCS1b rescues AβO-induced impairment of NRX-mediated presynaptic organization and presynaptic vesicle recycling and AβO-induced structural defects in excitatory synapses. Thus, our data suggest a role for SorCS1 in the rescue of AβO-induced NRX dysfunction and synaptic pathology, providing the basis for a novel potential therapeutic strategy for Alzheimer's disease.
Collapse
Affiliation(s)
- Alfred Kihoon Lee
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Nayoung Yi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Husam Khaled
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Benjamin Feller
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Hideto Takahashi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
| |
Collapse
|
35
|
Pavon MV, Navakkode S, Wong LW, Sajikumar S. Inhibition of Nogo-A rescues synaptic plasticity and associativity in APP/PS1 animal model of Alzheimer's disease. Semin Cell Dev Biol 2023; 139:111-120. [PMID: 35431138 DOI: 10.1016/j.semcdb.2022.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and cognitive decline. Synaptic impairment is one of the first events to occur in the progression of this disease. Synaptic plasticity and cellular association of various plastic events have been shown to be affected in AD models. Nogo-A, a well-known axonal growth inhibitor with a recently discovered role as a plasticity suppressor, and its main receptor Nogo-66 receptor 1 (NGR1) have been found to be overexpressed in the hippocampus of Alzheimer's patients. However, the role of Nogo-A and its receptor in the pathology of AD is still widely unknown. In this work we set out to investigate whether Nogo-A is working as a plasticity suppressor in AD. Our results show that inhibition of the Nogo-A pathway via the Nogo-R antibody in an Alzheimer's mouse model, APP/PS1, leads to the restoration of both synaptic plasticity and associativity in a protein synthesis and NMDR-dependent manner. We also show that inhibition of the p75NTR pathway, which is strongly associated with NGR1, restores synaptic plasticity as well. Mechanistically, we propose that the restoration of synaptic plasticity in APP/PS1 via inhibition of the Nogo-A pathway is due to the modulation of the RhoA-ROCK2 pathway and increase in plasticity related proteins. Our study identifies Nogo-A as a plasticity suppressor in AD models hence targeting Nogo-A could be a promising strategy to understanding AD pathology.
Collapse
Affiliation(s)
- Maria Vazquez Pavon
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Sheeja Navakkode
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Lik-Wei Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore; Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore.
| |
Collapse
|
36
|
Chua JJE. HEBP1 - An early trigger for neuronal cell death and circuit dysfunction in Alzheimer's disease. Semin Cell Dev Biol 2023; 139:102-110. [PMID: 35842370 DOI: 10.1016/j.semcdb.2022.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 07/05/2022] [Accepted: 07/05/2022] [Indexed: 12/31/2022]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder that gradually impairs memory, cognition and the ability to perform simple daily tasks. It is the most prevalent form of dementia in the elderly and its incidence increases exponentially with age. Neuronal and synapse loss, key hallmarks of the disorder, are widely regarded to occur early during the onset of AD, and the extent of this loss closely correlates with the progression of cognitive decline and dysfunction of the underlying neuronal circuity. Nevertheless, the mechanisms driving neuronal and synapse loss during early AD remains poorly understood. This review focuses on Heme-binding protein 1 (HEBP1), a mitochondrial-associated protein that has recently emerged as an important mediator of neuronal cell death during early AD pathogenesis. Acting downstream of Aβ and heme, HEBP1-mediated apoptosis contributes to neuronal loss and neuronal circuit dysfunction. Deleting HEBP1 expression in neurons protects them from heme- and Aβ-induced apoptosis, both of which are mechanisms implicated in neurodegeneration. HEBP1 participates in heme metabolism and binds to heme to modulate mitochondrial dynamics vital to the maintenance of neural circuitry that is affected in AD. HEBP1 elevation is also associated with AGE/RAGE-related neuronal damage, further implicating its involvement in neuronal loss during early AD. Moreover, F2L, a cleavage product of HEBP1 modulates inflammation. Collectively, these findings highlight the importance of HEBP1 in the disruption of neural circuits during early AD.
Collapse
Affiliation(s)
- John Jia En Chua
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; LSI Neurobiology Programme, National University of Singapore, Singapore; Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Health Innovation and Technology, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A⁎STAR), Singapore.
| |
Collapse
|
37
|
Maghsoodi F, Martin TD, Chi EY. Partial Destabilization of Amyloid-β Protofibril by Methionine Photo-Oxidation: A Molecular Dynamic Simulation Study. ACS OMEGA 2023; 8:10148-10159. [PMID: 36969430 PMCID: PMC10035002 DOI: 10.1021/acsomega.2c07468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
Selective photosensitized oxidation of amyloid protein aggregates is being investigated as a possible therapeutic strategy for treating Alzheimer's disease (AD). Photo-oxidation has been shown to degrade amyloid-β (Aβ) aggregates and ameliorate aggregate toxicity in vitro and reduce aggregate levels in the brains of AD animal models. To shed light on the mechanism by which photo-oxidation induces fibril destabilization, we carried out an all-atom molecular dynamics (MD) simulation to examine the effect of methionine (Met35) oxidation on the conformation and stability of a β-sheet-rich Aβ9-40 protofibril. Analyses of up to 1 μs simulations showed that the oxidation of the Met35 residues, which resulted in the addition of hydrophilic oxygens in the fibril core, reduced the overall conformational stability of the protofibril. Specifically, Met35 disrupted the hydrophobic interface that stabilizes the stacking of the two hexamers that comprise the protofibril. The oxidized protofibril is more solvent exposed and exhibits more backbone flexibility. However, the protofibril retained the underlying U-shaped architecture of each peptide upon oxidation, and although some loss of β-sheets occurred, a significant portion remained. Our simulation results are thus consistent with our experimental observation that photo-oxidation of Aβ40 fibril resulted in the dis-agglomeration and fragmentation of Aβ fibrils but did not cause complete disruption of the fibrillar morphology or β-sheet structures. The partial destabilization of Aβ aggregates supports the further development of photosensitized platforms for the targeting and clearing of Aβ aggregates as a therapeutic strategy for treating AD.
Collapse
Affiliation(s)
- Fahimeh Maghsoodi
- Nanoscience
and Microsystems Engineering Graduate Program, University of New Mexico, Albuquerque, New Mexico 87131, United States
- Center
for Biomedical Engineering, University of
New Mexico, Albuquerque, New Mexico 87131, United States
| | - Tye D. Martin
- Center
for Biomedical Engineering, University of
New Mexico, Albuquerque, New Mexico 87131, United States
| | - Eva Y. Chi
- Center
for Biomedical Engineering, University of
New Mexico, Albuquerque, New Mexico 87131, United States
- Department
of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| |
Collapse
|
38
|
AdipoRon mitigates tau pathology and restores mitochondrial dynamics via AMPK-related pathway in a mouse model of Alzheimer's disease. Exp Neurol 2023; 363:114355. [PMID: 36868546 DOI: 10.1016/j.expneurol.2023.114355] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/13/2023] [Accepted: 02/18/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a complicated and refractory neurodegenerative disease that is typically characterized by memory loss and multiple cognitive impairments. Multiple neuropathology including hyperphosphorylated tau formation and accumulation, dysregulated mitochondrial dynamics, and synaptic damage have been well implicated in the progression of AD. So far, there are few valid and effective therapeutic modalities for treatment. AdipoRon, a specific adiponectin (APN) receptor agonist, is reported to be associated with cognitive deficits improvement. In the present study, we attempt to explore the potential therapeutic effects of AdipoRon on tauopathy and related molecular mechanisms. METHODS In this study, P301S tau transgenic mice were used. The plasma level of APN was detected by ELISA. The level of APN receptors was qualified by western blot and immunofluorescence. 6-month-old mice were treated with AdipoRon or vehicle by oral administration daily for 4 months. The benefits of AdipoRon on tau hyperphosphorylation, mitochondrial dynamics, and synaptic function were detected by western blot, immunohistochemistry, immunofluorescence, Golgi staining and transmission electron microscopy. Morris water maze test and novel object recognition test were conducted to explore memory impairments. RESULTS Compared with wild-type mice, the expression of APN in plasma in 10-month-old P301S mice was obviously decreased. APN receptors in the hippocampus were increased in the hippocampus. AdipoRon treatment significantly rescued memory deficits in P301S mice. Besides, AdipoRon treatment was also detected to improve synaptic function, enhance mitochondrial fusion, and mitigate hyperphosphorylated tau accumulation in P301S mice and SY5Y cells. Mechanistically, AMPK/SIRT3 and AMPK/GSK3β signaling pathways are demonstrated to be involved in AdipoRon-mediated benefits on mitochondrial dynamics and tau accumulation, respectively, and inhibition of AMPK related pathways showed counteracted effects. CONCLUSION Our results demonstrated that AdipoRon treatment could significantly mitigate tau pathology, improve synaptic damage, and restore mitochondrial dynamics via the AMPK-related pathway, which provides a novel potential therapeutic approach to retard the progression of AD and other tauopathies diseases.
Collapse
|
39
|
Kim S, Nam Y, Kim MJ, Kwon SH, Jeon J, Shin SJ, Park S, Chang S, Kim HU, Lee YY, Kim HS, Moon M. Proteomic analysis for the effects of non-saponin fraction with rich polysaccharide from Korean Red Ginseng on Alzheimer's disease in a mouse model. J Ginseng Res 2023; 47:302-310. [PMID: 36926613 PMCID: PMC10014184 DOI: 10.1016/j.jgr.2022.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022] Open
Abstract
Background The most common type of dementia, Alzheimer's disease (AD), is marked by the formation of extracellular amyloid beta (Aβ) plaques. The impairments of axons and synapses appear in the process of Aβ plaques formation, and this damage could cause neurodegeneration. We previously reported that non-saponin fraction with rich polysaccharide (NFP) from Korean Red Ginseng (KRG) showed neuroprotective effects in AD. However, precise molecular mechanism of the therapeutic effects of NFP from KRG in AD still remains elusive. Methods To investigate the therapeutic mechanisms of NFP from KRG on AD, we conducted proteomic analysis for frontal cortex from vehicle-treated wild-type, vehicle-treated 5XFAD mice, and NFP-treated 5XFAD mice by using nano-LC-ESI-MS/MS. Metabolic network analysis was additionally performed as the effects of NFP appeared to be associated with metabolism according to the proteome analysis. Results Starting from 5,470 proteins, 2,636 proteins were selected for hierarchical clustering analysis, and finally 111 proteins were further selected for protein-protein interaction network analysis. A series of these analyses revealed that proteins associated with synapse and mitochondria might be linked to the therapeutic mechanism of NFP. Subsequent metabolic network analysis via genome-scale metabolic models that represent the three mouse groups showed that there were significant changes in metabolic fluxes of mitochondrial carnitine shuttle pathway and mitochondrial beta-oxidation of polyunsaturated fatty acids. Conclusion Our results suggested that the therapeutic effects of NFP on AD were associated with synaptic- and mitochondrial-related pathways, and they provided targets for further rigorous studies on precise understanding of the molecular mechanism of NFP.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea.,Research Institute for Dementia Science, Konyang University, Daejeon, Republic of Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Min-Jeong Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Seung-Hyun Kwon
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Junhyeok Jeon
- Department of Chemical and Biomolecular Engineering (BK21 four), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Soyoon Park
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, California, United States
| | - Sungjae Chang
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Hyun Uk Kim
- Department of Chemical and Biomolecular Engineering (BK21 four), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Yong Yook Lee
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Hak Su Kim
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea.,Research Institute for Dementia Science, Konyang University, Daejeon, Republic of Korea
| |
Collapse
|
40
|
Loss of brain energy metabolism control as a driver for memory impairment upon insulin resistance. Biochem Soc Trans 2023; 51:287-301. [PMID: 36606696 DOI: 10.1042/bst20220789] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023]
Abstract
The pathophysiological mechanisms intersecting metabolic and neurodegenerative disorders include insulin resistance, which has a strong involvement of environmental factors. Besides central regulation of whole-body homeostasis, insulin in the central nervous system controls molecular signalling that is critical for cognitive performance, namely signalling through pathways that modulate synaptic transmission and plasticity, and metabolism in neurons and astrocytes. This review provides an overview on how insulin signalling in the brain might regulate brain energy metabolism, and further identified molecular mechanisms by which brain insulin resistance might impair synaptic fuelling, and lead to cognitive deterioration.
Collapse
|
41
|
Li S. The β-adrenergic hypothesis of synaptic and microglial impairment in Alzheimer's disease. J Neurochem 2023; 165:289-302. [PMID: 36799441 DOI: 10.1111/jnc.15782] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease originating partly from amyloid β protein-induced synaptic failure. As damaging of noradrenergic neurons in the locus coeruleus (LC) occurs at the prodromal stage of AD, activation of adrenergic receptors could serve as the first line of defense against the onset of the disease. Activation of β2 -ARs strengthens long-term potentiation (LTP) and synaptic activity, thus improving learning and memory. Physical stimulation of animals exposed to an enriched environment (EE) leads to the activation of β2 -ARs and prevents synaptic dysfunction. EE also suppresses neuroinflammation, suggesting that β2 -AR agonists may play a neuroprotective role. The β2 -AR agonists used for respiratory diseases have been shown to have an anti-inflammatory effect. Epidemiological studies further support the beneficial effects of β2 -AR agonists on several neurodegenerative diseases. Thus, I propose that β2 -AR agonists may provide therapeutic value in combination with novel treatments for AD.
Collapse
Affiliation(s)
- Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Glebov OO, Williamson D, Owen DM, Hortobágyi T, Troakes C, Aarsland D. Structural synaptic signatures of Alzheimer's disease and dementia with Lewy bodies in the male brain. Neuropathol Appl Neurobiol 2023; 49:e12852. [PMID: 36181001 PMCID: PMC10092423 DOI: 10.1111/nan.12852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Oleg O Glebov
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China.,Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - David Williamson
- Randall Centre for Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Dylan M Owen
- Institute of Immunology and Immunotherapy, School of Mathematics and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Tibor Hortobágyi
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,ELKH-DE Cerebrovascular and Neurodegenerative Research Group and Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Claire Troakes
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
43
|
Colussi C, Aceto G, Ripoli C, Bertozzi A, Li Puma DD, Paccosi E, D'Ascenzo M, Grassi C. Cytoplasmic HDAC4 recovers synaptic function in the 3×Tg mouse model of Alzheimer's disease. Neuropathol Appl Neurobiol 2023; 49:e12861. [PMID: 36331820 PMCID: PMC10099707 DOI: 10.1111/nan.12861] [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: 05/24/2022] [Revised: 08/26/2022] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
Abstract
AIMS Early dysfunction in Alzheimer's disease (AD) is characterised by alterations of synapse structure and function leading to dysmorphic neurites, decreased spine density, impaired synaptic plasticity and cognitive deficits. The class II member HDAC4, which recently emerged as a crucial factor in shaping synaptic plasticity and memory, was found to be altered in AD. We investigated how the modulation of HDAC4 may contribute to counteracting AD pathogenesis. METHODS Using a cytoplasmic HDAC4 mutant (HDAC4SD ), we studied the recovery of synaptic function in hippocampal tissue and primary neurons from the triple-transgenic mouse model of AD (3×Tg-AD). RESULTS Here, we report that in wild-type mice, HDAC4 is localised at synapses and interacts with postsynaptic proteins, whereas in the 3×Tg-AD, it undergoes nuclear import, reducing its interaction with synaptic proteins. Of note, HDAC4 delocalisation was induced by both amyloid-β and tau accumulation. Overexpression of the HDAC4SD mutant in CA1 pyramidal neurons of organotypic hippocampal slices obtained from 3×Tg-AD mice increased dendritic length and promoted the enrichment of N-cadherin, GluA1, PSD95 and CaMKII proteins at the synaptic level compared with AD neurons transfected with the empty vector. Moreover, HDAC4 overexpression recovered the level of SUMO2/3ylation of PSD95 in AD hippocampal tissue, and in AD organotypic hippocampal slices, the HDAC4SD rescued spine density and synaptic transmission. CONCLUSIONS These results highlight a new role of cytoplasmic HDAC4 in providing a structural and enzymatic regulation of postsynaptic proteins. Our findings suggest that controlling HDAC4 localisation may represent a promising strategy to rescue synaptic function in AD, potentially leading to memory improvement.
Collapse
Affiliation(s)
- Claudia Colussi
- Department of Engineering, Istituto di Analisi dei Sistemi ed Informatica 'Antonio Ruberti', National Research Council, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giuseppe Aceto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alessia Bertozzi
- Department of Engineering, Istituto di Analisi dei Sistemi ed Informatica 'Antonio Ruberti', National Research Council, Rome, Italy
| | - Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Elena Paccosi
- Department of Engineering, Istituto di Analisi dei Sistemi ed Informatica 'Antonio Ruberti', National Research Council, Rome, Italy
| | - Marcello D'Ascenzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
44
|
Liu S, Zhang Z, Li L, Yao L, Ma Z, Li J. ADAM10- and γ-secretase-dependent cleavage of the transmembrane protein PTPRT attenuates neurodegeneration in the mouse model of Alzheimer's disease. FASEB J 2023; 37:e22734. [PMID: 36583697 DOI: 10.1096/fj.202201396r] [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: 08/25/2022] [Revised: 11/20/2022] [Accepted: 12/14/2022] [Indexed: 12/31/2022]
Abstract
PTPRT (receptor-type tyrosine-protein phosphatase T), a brain-specific type 1 transmembrane protein, plays an important role in neurodevelopment and synapse formation. However, whether abnormal PTPRT signaling is associated with Alzheimer's disease (AD) remains elusive. Here, we report that Ptprt mRNA expression is found to be downregulated in the brains of both human and mouse models of AD. We further identified that the PTPRT intracellular domain (PICD), which is released by ADAM10- and γ-secretase-dependent cleavage of PTPRT, efficiently translocates to the nucleus via a conserved nuclear localization signal (NLS). We show that inhibition of nuclear translocation of PICD leads to an accumulation of phosphorylated signal transducer and activator of transcription 3 (pSTAT3), a substrate of PTPRT-eventually resulting in neuronal cell death. Consistently, RNA sequencing reveals that overexpression of PICD leads to changes in the expression of genes that are functionally associated with synapse formation, cell adhesion, and protein dephosphorylation. Moreover, overexpression of PICD not only decreases the level of phospho-STAT3Y705 and amyloid β production in the hippocampus of APP/PS1 mice but also partially improves synaptic function and behavioral deficits in this mouse model of AD. These findings suggest that a novel role of the ADAM 10- and γ-secretase-dependent cleavage of PTPRT may alleviate the AD-like neurodegenerative processes.
Collapse
Affiliation(s)
- Siling Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Zhongyu Zhang
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Lianwei Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Li Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Zhanshan Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jiali Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,National Institute on Drug Dependence, Peking University, Beijing, China.,IDG/McGovern Institute for Brain Research, Peking University, Beijing, China.,Kunming Primate Research Center of the Chinese Academy of Sciences, Kunming, China.,National Research Facility for Phenotypic and Genetic Analysis of Model Animals, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
45
|
Warpechowski M, Warpechowski J, Kulczyńska-Przybik A, Mroczko B. Biomarkers of Activity-Dependent Plasticity and Persistent Enhancement of Synaptic Transmission in Alzheimer Disease: A Review of the Current Status. Med Sci Monit 2023; 29:e938826. [PMID: 36600577 PMCID: PMC9832729 DOI: 10.12659/msm.938826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Alzheimer disease (AD) is a chronic and heterogeneous neurodegenerative disorder characterized by complex pathological processes involving neuroinflammation, neurodegeneration, and synaptic dysfunction. Understanding the exact neurobiological mechanisms underlying AD pathology may help to provide a biomarker for early diagnosis or at least for assessment of vulnerability to dementia development. Neural plasticity is defined as a capability of the brain to respond to alterations including aging, injury, or learning, with a crucial role of synaptic elements. Long-term potentiation (LTP) and long-term depression (LTD) are important in regulating synaptic connections between neural cells in functional plasticity. Synaptic loss and impairment of the brain's plasticity in AD leads to cognitive impairment, and one of important roles of synaptic biomarkers is monitoring synaptic dysfunction, response to treatment, and predicting future development of AD. Synaptic biomarkers are undoubtedly very promising in developing novel approach to AD treatment and control, especially in the era of aging of societies, which is one of the most common risk factor of AD. Implementing a widespread measurement of synaptic biomarkers of AD will probably be crucial in early diagnosis of AD, early therapeutic intervention, monitoring progression of the disease, or response to treatment. One of the most important challenges is finding a biomarker whose blood concentration correlates with its level in the central nervous system (CNS). This review aims to present the current status of biomarkers of activity-dependent plasticity and persistent enhancement of synaptic transmission in Alzheimer disease.
Collapse
Affiliation(s)
- Marcin Warpechowski
- Department of Statistics and Medical Informatics, Medical University of Białystok, Białystok, Poland
| | | | | | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, Białystok, Poland,Department of Biochemical Diagnostics, University Hospital of Białystok, Białystok, Poland
| |
Collapse
|
46
|
de Veij Mestdagh CF, Koopmans F, Breiter JC, Timmerman JA, Vogelaar PC, Krenning G, Mansvelder HD, Smit AB, Henning RH, van Kesteren RE. The hibernation-derived compound SUL-138 shifts the mitochondrial proteome towards fatty acid metabolism and prevents cognitive decline and amyloid plaque formation in an Alzheimer's disease mouse model. Alzheimers Res Ther 2022; 14:183. [PMID: 36482297 PMCID: PMC9733344 DOI: 10.1186/s13195-022-01127-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide and remains without effective cure. Increasing evidence is supporting the mitochondrial cascade hypothesis, proposing that loss of mitochondrial fitness and subsequent ROS and ATP imbalance are important contributors to AD pathophysiology. METHODS Here, we tested the effects of SUL-138, a small hibernation-derived molecule that supports mitochondrial bioenergetics via complex I/IV activation, on molecular, physiological, behavioral, and pathological outcomes in APP/PS1 and wildtype mice. RESULTS SUL-138 treatment rescued long-term potentiation and hippocampal memory impairments and decreased beta-amyloid plaque load in APP/PS1 mice. This was paralleled by a partial rescue of dysregulated protein expression in APP/PS1 mice as assessed by mass spectrometry-based proteomics. In-depth analysis of protein expression revealed a prominent effect of SUL-138 in APP/PS1 mice on mitochondrial protein expression. SUL-138 increased the levels of proteins involved in fatty acid metabolism in both wildtype and APP/PS1 mice. Additionally, in APP/PS1 mice only, SUL-138 increased the levels of proteins involved in glycolysis and amino acid metabolism pathways, indicating that SUL-138 rescues mitochondrial impairments that are typically observed in AD. CONCLUSION Our study demonstrates a SUL-138-induced shift in metabolic input towards the electron transport chain in synaptic mitochondria, coinciding with increased synaptic plasticity and memory. In conclusion, targeting mitochondrial bioenergetics might provide a promising new way to treat cognitive impairments in AD and reduce disease progression.
Collapse
Affiliation(s)
- Christina F. de Veij Mestdagh
- grid.12380.380000 0004 1754 9227Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands ,grid.4494.d0000 0000 9558 4598Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, the Netherlands ,grid.16872.3a0000 0004 0435 165XAlzheimer Center Amsterdam, Vrije Universiteit Amsterdam and Amsterdam UMC location VUmc , Amsterdam, The Netherlands
| | - Frank Koopmans
- grid.12380.380000 0004 1754 9227Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Jonathan C. Breiter
- grid.4494.d0000 0000 9558 4598Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, the Netherlands
| | - Jaap A. Timmerman
- grid.12380.380000 0004 1754 9227Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Pieter C. Vogelaar
- grid.4494.d0000 0000 9558 4598Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, the Netherlands ,Sulfateq B.V., Groningen, The Netherlands
| | - Guido Krenning
- grid.4494.d0000 0000 9558 4598Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, the Netherlands ,Sulfateq B.V., Groningen, The Netherlands ,grid.4494.d0000 0000 9558 4598Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| | - Huibert D. Mansvelder
- grid.12380.380000 0004 1754 9227Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - August B. Smit
- grid.12380.380000 0004 1754 9227Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Robert H. Henning
- grid.4494.d0000 0000 9558 4598Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, the Netherlands
| | - Ronald E. van Kesteren
- grid.12380.380000 0004 1754 9227Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
47
|
Lan G, Li A, Liu Z, Ma S, Guo T. Presynaptic membrane protein dysfunction occurs prior to neurodegeneration and predicts faster cognitive decline. Alzheimers Dement 2022. [DOI: 10.1002/alz.12890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Guoyu Lan
- Institute of Biomedical Engineering Shenzhen Bay Laboratory Shenzhen China
- Tsinghua Shenzhen International Graduate School (SIGS) Tsinghua University Shenzhen China
| | - Anqi Li
- Institute of Biomedical Engineering Shenzhen Bay Laboratory Shenzhen China
| | - Zhen Liu
- Institute of Biomedical Engineering Shenzhen Bay Laboratory Shenzhen China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS) Tsinghua University Shenzhen China
| | - Tengfei Guo
- Institute of Biomedical Engineering Shenzhen Bay Laboratory Shenzhen China
- Institute of Biomedical Engineering Peking University Shenzhen Graduate School Shenzhen China
| | | |
Collapse
|
48
|
Presenilin and APP Regulate Synaptic Kainate Receptors. J Neurosci 2022; 42:9253-9262. [PMID: 36288945 PMCID: PMC9761675 DOI: 10.1523/jneurosci.0297-22.2022] [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: 02/10/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 02/02/2023] Open
Abstract
Kainate receptors (KARs) form a family of ionotropic glutamate receptors that regulate the activity of neuronal networks by both presynaptic and postsynaptic mechanisms. Their implication in pathologies is well documented for epilepsy. The higher prevalence of epileptic symptoms in Alzheimer's disease (AD) patients questions the role of KARs in AD. Here we investigated whether the synaptic expression and function of KARs was impaired in mouse models of AD. We addressed this question by immunostaining and electrophysiology at synapses between mossy fibers and CA3 pyramidal cells, in which KARs are abundant and play a prominent physiological role. We observed a decrease of the immunostaining for GluK2 in the stratum lucidum in CA3, and of the amplitude and decay time of synaptic currents mediated by GluK2-containing KARs in an amyloid mouse model (APP/PS1) of AD. Interestingly, a similar phenotype was observed in CA3 pyramidal cells in male and female mice with a genetic deletion of either presenilin or APP/APLP2 as well as in organotypic cultures treated with γ-secretase inhibitors. Finally, the GluK2 protein interacts with full-length and C-terminal fragments of APP. Overall, our data suggest that APP stabilizes KARs at synapses, possibly through a transsynaptic mechanism, and this interaction is under the control the γ-secretase proteolytic activity of presenilin.SIGNIFICANCE STATEMENT Synaptic impairment correlates strongly with cognitive deficits in Alzheimer's disease (AD). In this context, many studies have addressed the dysregulation of AMPA and NMDA ionotropic glutamate receptors. Kainate receptors (KARs), which form the third family of iGluRs, represent an underestimated actor in the regulation of neuronal circuits and have not yet been examined in the context of AD. Here we provide evidence that synaptic KARs are markedly impaired in a mouse model of AD. Additional experiments indicate that the γ-secretase activity of presenilin acting on the amyloid precursor protein controls synaptic expression of KAR. This study clearly indicates that KARs should be taken into consideration whenever addressing synaptic dysfunction and related cognitive deficits in the context of AD.
Collapse
|
49
|
van Nifterick AM, Gouw AA, van Kesteren RE, Scheltens P, Stam CJ, de Haan W. A multiscale brain network model links Alzheimer’s disease-mediated neuronal hyperactivity to large-scale oscillatory slowing. Alzheimers Res Ther 2022; 14:101. [PMID: 35879779 PMCID: PMC9310500 DOI: 10.1186/s13195-022-01041-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 07/02/2022] [Indexed: 01/30/2023]
Abstract
Background Neuronal hyperexcitability and inhibitory interneuron dysfunction are frequently observed in preclinical animal models of Alzheimer’s disease (AD). This study investigates whether these microscale abnormalities explain characteristic large-scale magnetoencephalography (MEG) activity in human early-stage AD patients. Methods To simulate spontaneous electrophysiological activity, we used a whole-brain computational network model comprised of 78 neural masses coupled according to human structural brain topology. We modified relevant model parameters to simulate six literature-based cellular scenarios of AD and compare them to one healthy and six contrast (non-AD-like) scenarios. The parameters include excitability, postsynaptic potentials, and coupling strength of excitatory and inhibitory neuronal populations. Whole-brain spike density and spectral power analyses of the simulated data reveal mechanisms of neuronal hyperactivity that lead to oscillatory changes similar to those observed in MEG data of 18 human prodromal AD patients compared to 18 age-matched subjects with subjective cognitive decline. Results All but one of the AD-like scenarios showed higher spike density levels, and all but one of these scenarios had a lower peak frequency, higher spectral power in slower (theta, 4–8Hz) frequencies, and greater total power. Non-AD-like scenarios showed opposite patterns mainly, including reduced spike density and faster oscillatory activity. Human AD patients showed oscillatory slowing (i.e., higher relative power in the theta band mainly), a trend for lower peak frequency and higher total power compared to controls. Combining model and human data, the findings indicate that neuronal hyperactivity can lead to oscillatory slowing, likely due to hyperexcitation (by hyperexcitability of pyramidal neurons or greater long-range excitatory coupling) and/or disinhibition (by reduced excitability of inhibitory interneurons or weaker local inhibitory coupling strength) in early AD. Conclusions Using a computational brain network model, we link findings from different scales and models and support the hypothesis of early-stage neuronal hyperactivity underlying E/I imbalance and whole-brain network dysfunction in prodromal AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-01041-4.
Collapse
|
50
|
Avraham O, Chamessian A, Feng R, Yang L, Halevi AE, Moore AM, Gereau RW, Cavalli V. Profiling the molecular signature of satellite glial cells at the single cell level reveals high similarities between rodents and humans. Pain 2022; 163:2348-2364. [PMID: 35503034 PMCID: PMC9522926 DOI: 10.1097/j.pain.0000000000002628] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT Peripheral sensory neurons located in dorsal root ganglia relay sensory information from the peripheral tissue to the brain. Satellite glial cells (SGCs) are unique glial cells that form an envelope completely surrounding each sensory neuron soma. This organization allows for close bidirectional communication between the neuron and its surrounding glial coat. Morphological and molecular changes in SGC have been observed in multiple pathological conditions such as inflammation, chemotherapy-induced neuropathy, viral infection, and nerve injuries. There is evidence that changes in SGC contribute to chronic pain by augmenting the neuronal activity in various rodent pain models. Satellite glial cells also play a critical role in axon regeneration. Whether findings made in rodent model systems are relevant to human physiology have not been investigated. Here, we present a detailed characterization of the transcriptional profile of SGC in mice, rats, and humans at the single cell level. Our findings suggest that key features of SGC in rodent models are conserved in humans. Our study provides the potential to leverage rodent SGC properties and identify potential targets in humans for the treatment of nerve injuries and alleviation of painful conditions.
Collapse
Affiliation(s)
- Oshri Avraham
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Alexander Chamessian
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Rui Feng
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Lite Yang
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Neuroscience Program, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Alexandra E. Halevi
- Department of Plastic and Reconstructive Surgery, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Amy M. Moore
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus Ohio, USA
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|