1
|
Gómez-Pascual A, Rocamora-Pérez G, Ibanez L, Botía JA. Targeted co-expression networks for the study of traits. Sci Rep 2024; 14:16675. [PMID: 39030261 PMCID: PMC11271532 DOI: 10.1038/s41598-024-67329-7] [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: 12/28/2023] [Accepted: 07/10/2024] [Indexed: 07/21/2024] Open
Abstract
Weighted Gene Co-expression Network Analysis (WGCNA) is a widely used approach for the generation of gene co-expression networks. However, networks generated with this tool usually create large modules with a large set of functional annotations hard to decipher. We have developed TGCN, a new method to create Targeted Gene Co-expression Networks. This method identifies the transcripts that best predict the trait of interest based on gene expression using a refinement of the LASSO regression. Then, it builds the co-expression modules around those transcripts. Algorithm properties were characterized using the expression of 13 brain regions from the Genotype-Tissue Expression project. When comparing our method with WGCNA, TGCN networks lead to more precise modules that have more specific and yet rich biological meaning. Then, we illustrate its applicability by creating an APP-TGCN on The Religious Orders Study and Memory and Aging Project dataset, aiming to identify the molecular pathways specifically associated with APP role in Alzheimer's disease. Main biological findings were further validated in two independent cohorts. In conclusion, we provide a new framework that serves to create targeted networks that are smaller, biologically relevant and useful in high throughput hypothesis driven research. The TGCN R package is available on Github: https://github.com/aliciagp/TGCN .
Collapse
Affiliation(s)
- A Gómez-Pascual
- Communications Engineering and Information Department, University of Murcia, 30100, Murcia, Spain
| | - G Rocamora-Pérez
- Department of Genetics and Genomic Medicine Research and Teaching, UCL GOS Institute of Child Health, London, WC1N 1EH, UK
| | - L Ibanez
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - J A Botía
- Communications Engineering and Information Department, University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
2
|
Quintela-López T, Lezmy J. Homeostatic plasticity of axonal excitable sites in Alzheimer's disease. Front Neurosci 2023; 17:1277251. [PMID: 37937068 PMCID: PMC10626477 DOI: 10.3389/fnins.2023.1277251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Affiliation(s)
| | - Jonathan Lezmy
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
3
|
Furube E, Ohgidani M, Yoshida S. Systemic Inflammation Leads to Changes in the Intracellular Localization of KLK6 in Oligodendrocytes in Spinal Cord White Matter. Neurochem Res 2023; 48:2645-2659. [PMID: 37067738 DOI: 10.1007/s11064-023-03929-5] [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: 11/21/2022] [Revised: 02/24/2023] [Accepted: 03/31/2023] [Indexed: 04/18/2023]
Abstract
Axonal injury and demyelination occur in demyelinating diseases, such as multiple sclerosis, and the detachment of myelin from axons precedes its degradation. Paranodes are the areas at which each layer of the myelin sheath adheres tightly to axons. The destruction of nodal and paranodal structures during inflammation is an important pathophysiology of various neurological disorders. However, the underlying pathological changes in these structures remain unclear. Kallikrein 6 (KLK6), a serine protease produced by oligodendrocytes, is involved in demyelinating diseases. In the present study, we intraperitoneally injected mice with LPS for several days and examined changes in the localization of KLK6. Transient changes in the intracellular localization of KLK6 to paranodes in the spinal cord were observed during LPS-induced systemic inflammation. However, these changes were not detected in the upper part of brain white matter. LPS-induced changes were suppressed by minocycline, suggesting the involvement of microglia. Moreover, nodal lengths were elongated in LPS-treated wild-type mice, but not in LPS-treated KLK6-KO mice. These results demonstrate the potential involvement of KLK6 in the process of demyelination.
Collapse
Affiliation(s)
- Eriko Furube
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan.
| | - Masahiro Ohgidani
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan
| | - Shigetaka Yoshida
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan
| |
Collapse
|
4
|
Dolma S, Joshi A. The Node of Ranvier as an Interface for Axo-Glial Interactions: Perturbation of Axo-Glial Interactions in Various Neurological Disorders. J Neuroimmune Pharmacol 2023; 18:215-234. [PMID: 37285016 DOI: 10.1007/s11481-023-10072-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 05/19/2023] [Indexed: 06/08/2023]
Abstract
The action potential conduction along the axon is highly dependent on the healthy interactions between the axon and myelin-producing glial cells. Myelin, which facilitates action potential, is the protective insulation around the axon formed by Schwann cells and oligodendrocytes in the peripheral (PNS) and central nervous system (CNS), respectively. Myelin is a continuous structure with intermittent gaps called nodes of Ranvier, which are the sites enriched with ion channels, transmembrane, scaffolding, and cytoskeletal proteins. Decades-long extensive research has identified a comprehensive proteome with strictly regularized localization at the node of Ranvier. Concurrently, axon-glia interactions at the node of Ranvier have gathered significant attention as the pathophysiological targets for various neurodegenerative disorders. Numerous studies have shown the alterations in the axon-glia interactions culminating in neurological diseases. In this review, we have provided an update on the molecular composition of the node of Ranvier. Further, we have discussed in detail the consequences of disruption of axon-glia interactions during the pathogenesis of various CNS and PNS disorders.
Collapse
Affiliation(s)
- Sonam Dolma
- Department of Pharmacy, Birla Institute of Technology and Sciences- Pilani, Hyderabad campus, Telangana state, India
| | - Abhijeet Joshi
- Department of Pharmacy, Birla Institute of Technology and Sciences- Pilani, Hyderabad campus, Telangana state, India.
| |
Collapse
|
5
|
Jiang R, Chi XD, Jing Y, Wang B, Li S. Reduction of NgR in perforant path protects neuronal morphology and function in APP/PS1 transgenic mice. Aging (Albany NY) 2023; 15:2158-2169. [PMID: 36961417 PMCID: PMC10085588 DOI: 10.18632/aging.204605] [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: 12/16/2021] [Accepted: 03/23/2022] [Indexed: 03/25/2023]
Abstract
Neuronal loss is the central abnormality occurring in brains suffering from Alzheimer's disease (AD). The notion that AD causes the death of neurons point towards protection of neuronal morphology and function as important therapeutic strategies. The perforant path projections from the entorhinal cortex to the dentate gyrus is the most vulnerable circuit with respect to AD. It's known that the perforant path is a very important structure for synaptic plasticity and cognitive functions. NgR (Nogo receptor) is not only involved in limiting injury-induced axonal growth but also in pathological features of AD. So, the mechanism of how NgR affects the perforant path needs further investigation. In this study, the effect of NgR in the perforant path on the neuronal morphology and function in APP/PS1 transgenic mice was studied. The results showed that downregulation of NgR in perforant path ameliorate the damaged morphology and decreased number of neurons in APP/PS1 mice. Concurrently, NgR knockdown enhanced dendritic complexity and increased postsynaptic protein density in APP/PS1 mice. Furthermore, the RT-PCR results indicated that there is downregulation of M1 phenotypes of microglial gene expression in the hippocampus of TG-shNgR mice. Our study suggests that NgR plays a critical role in microglial phenotype polarization, which might account for the NgR knockdown in the perforant path initiated a decrease in neuronal death and improved synaptic function. Our study provided a better understanding of the perforant path and the role of NgR in AD pathogenesis, thus offering the potential application of hippocampal neurons in treatment of AD.
Collapse
Affiliation(s)
- Rong Jiang
- Department of Physiology, Binzhou Medical University, Yantai Campus, Yantai, China
| | - Xiao-Dong Chi
- Department of Neurology, Affiliated Dalian Friendship Hospital of Dalian Medical University, Dalian, China
| | - Yulong Jing
- Department of Traumatic Orthopedics, Yantaishan Hospital, Yantai, China
| | - Bin Wang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Shao Li
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| |
Collapse
|
6
|
Oxidative Stress as a Potential Mechanism Underlying Membrane Hyperexcitability in Neurodegenerative Diseases. Antioxidants (Basel) 2022; 11:antiox11081511. [PMID: 36009230 PMCID: PMC9405356 DOI: 10.3390/antiox11081511] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
Neurodegenerative diseases are characterized by gradually progressive, selective loss of anatomically or physiologically related neuronal systems that produce brain damage from which there is no recovery. Despite the differences in clinical manifestations and neuronal vulnerability, the pathological processes appear to be similar, suggesting common neurodegenerative pathways. It is well known that oxidative stress and the production of reactive oxygen radicals plays a key role in neuronal cell damage. It has been proposed that this stress, among other mechanisms, could contribute to neuronal degeneration and might be one of the factors triggering the development of these pathologies. Another common feature in most neurodegenerative diseases is neuron hyperexcitability, an aberrant electrical activity. This review, focusing mainly on primary motor cortex pyramidal neurons, critically evaluates the idea that oxidative stress and inflammation may be involved in neurodegeneration via their capacity to increase membrane excitability.
Collapse
|
7
|
Yuan D, Yang G, Wu W, Li Q, Xu D, Ntim M, Jiang C, Liu J, Zhang Y, Wang Y, Zhu D, Kundu S, Li A, Xiao Z, Ma Q, Li S. Reducing Nav1.6 expression attenuates the pathogenesis of Alzheimer's disease by suppressing BACE1 transcription. Aging Cell 2022; 21:e13593. [PMID: 35353937 PMCID: PMC9124306 DOI: 10.1111/acel.13593] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/12/2022] [Accepted: 03/07/2022] [Indexed: 12/14/2022] Open
Abstract
Aberrant increases in neuronal network excitability may contribute to cognitive deficits in Alzheimer's disease (AD). However, the mechanisms underlying hyperexcitability of neurons are not fully understood. Voltage‐gated sodium channels (VGSC or Nav), which are involved in the formation of excitable cell's action potential and can directly influence the excitability of neural networks, have been implicated in AD‐related abnormal neuronal hyperactivity and higher incidence of spontaneous non‐convulsive seizures. Here, we have shown that the reduction of VGSC α‐subunit Nav1.6 (by injecting adeno‐associated virus (AAV) with short hairpin RNA (shRNA) into the hippocampus) rescues cognitive impairments and attenuates synaptic deficits in APP/PS1 transgenic mice. Concurrently, amyloid plaques in the hippocampus and levels of soluble Aβ are significantly reduced. Interfering with Nav1.6 reduces the transcription level of β‐site APP‐cleaving enzyme 1 (BACE1), which is Aβ‐dependent. In the presence of Aβ oligomers, knockdown of Nav1.6 reduces intracellular calcium overload by suppressing reverse sodium–calcium exchange channel, consequently increasing inactive NFAT1 (the nuclear factor of activated T cells) levels and thus reducing BACE1 transcription. This mechanism leads to a reduction in the levels of Aβ in APP/PS1 transgenic mice, alleviates synaptic loss, improves learning and memory disorders in APP/PS1 mice after downregulating Nav1.6 in the hippocampus. Our study offers a new potential therapeutic strategy to counteract hippocampal hyperexcitability and subsequently rescue cognitive deficits in AD by selective blockade of Nav1.6 overexpression and/or hyperactivity.
Collapse
Affiliation(s)
- De‐Juan Yuan
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
- Department of Neurology and Clinical Research Center of Neurological Disease The Second Affiliated Hospital of Soochow University Suzhou China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Institute of Neuroscience Soochow University Suzhou China
- The Affiliated Wuxi No. 2 People’s Hospital of Nanjing Medical University Wuxi China
| | - Guang Yang
- Department of Thoracic Surgery Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Wei Wu
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
| | - Qi‐Fa Li
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
| | - De‐en Xu
- Department of Neurology and Clinical Research Center of Neurological Disease The Second Affiliated Hospital of Soochow University Suzhou China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Institute of Neuroscience Soochow University Suzhou China
- The Affiliated Wuxi No. 2 People’s Hospital of Nanjing Medical University Wuxi China
| | - Michael Ntim
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
| | - Chun‐Yan Jiang
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
| | - Ji‐Chuan Liu
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
- Department of Neurology and Clinical Research Center of Neurological Disease The Second Affiliated Hospital of Soochow University Suzhou China
| | - Yue Zhang
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
| | - Ying‐Zi Wang
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
| | - Dan‐Dan Zhu
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
| | - Supratik Kundu
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
| | - Ai‐Ping Li
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
| | - Zhi‐Cheng Xiao
- Development and Stem Cells Program Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology Monash University Melbourne Victoria Australia
| | - Quan‐Hong Ma
- Department of Neurology and Clinical Research Center of Neurological Disease The Second Affiliated Hospital of Soochow University Suzhou China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Institute of Neuroscience Soochow University Suzhou China
| | - Shao Li
- Department of Physiology College of Basic Medical Sciences Liaoning Provincial Key Laboratory of Cerebral Diseases National‐Local Joint Engineering Research Center for Drug‐Research and Development (R&D) of Neurodegenerative Diseases Dalian Medical University Dalian China
| |
Collapse
|
8
|
Kalle J, Pontus W, Lenka N, Simon S, Ann B, Gunnar B, Kaj B, Henrik Z, Markus A. Cerebrospinal fluid amyloid precursor protein as a potential biomarker of fatigue in multiple sclerosis: A pilot study. Mult Scler Relat Disord 2022; 63:103846. [DOI: 10.1016/j.msard.2022.103846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/15/2022] [Accepted: 05/01/2022] [Indexed: 11/28/2022]
|
9
|
An unbiased algorithm for objective separation of Alzheimer's, Alzheimer's mixed with cerebrovascular symptomology, and healthy controls from one another using electrovestibulography (EVestG). Med Biol Eng Comput 2022; 60:797-810. [PMID: 35102489 DOI: 10.1007/s11517-022-02507-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/18/2022] [Indexed: 10/19/2022]
Abstract
Diagnosis of Alzheimer's disease (AD) from AD with cerebrovascular disease pathology (AD-CVD) is a rising challenge. Using electrovestibulography (EVestG) measured signals, we develop an automated feature extraction and selection algorithm for an unbiased identification of AD and AD-CVD from healthy controls as well as their separation from each other. EVestG signals of 24 healthy controls, 16 individuals with AD, and 13 with AD-CVD were analyzed within two separate groupings: One-versus-One and One-versus-All. A multistage feature selection process was conducted over the training dataset using linear support vector machine (SVM) classification with 10-fold cross-validation, k nearest neighbors/averaging imputation, and exhaustive search. The most frequently selected features that achieved highest classification performance were selected. 10-fold cross-validation was applied via a linear SVM classification on the entire dataset. Multivariate analysis was run to test the between population differences while controlling for the covariates. Classification accuracies of ≥ 80% and 78% were achieved for the One-versus-All classification approach and AD versus AD-CVD separation, respectively. The results also held true after controlling for the effect of covariates. AD/AD-CVD participants showed smaller/larger EVestG averaged field potential signals compared to healthy controls and AD-CVD/AD participants. These characteristics are in line with our previous study results.
Collapse
|
10
|
Cheng S, Wang HN, Xu LJ, Li F, Miao Y, Lei B, Sun X, Wang Z. Soluble tumor necrosis factor-alpha-induced hyperexcitability contributes to retinal ganglion cell apoptosis by enhancing Nav1.6 in experimental glaucoma. J Neuroinflammation 2021; 18:182. [PMID: 34419081 PMCID: PMC8380326 DOI: 10.1186/s12974-021-02236-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/09/2021] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Neuroinflammation plays an important role in the pathogenesis of glaucoma. Tumor necrosis factor-alpha (TNF-α) is a major pro-inflammatory cytokine released from activated retinal glial cells in glaucoma. Here, we investigated how TNF-α induces retinal ganglion cell (RGC) hyperexcitability and injury. METHODS Whole-cell patch-clamp techniques were performed to explore changes in spontaneous firing and evoked action potentials, and Na+ currents in RGCs. Both intravitreal injection of TNF-α and chronic ocular hypertension (COH) models were used. Western blotting, immunofluorescence, quantitative real-time polymerase chain reaction (q-PCR), and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) techniques were employed to investigate the molecular mechanisms of TNF-α effects on RGCs. RESULTS Intravitreal injection of soluble TNF-α significantly increased the spontaneous firing frequencies of RGCs in retinal slices. When the synaptic transmissions were blocked, more than 90% of RGCs still showed spontaneous firing; both the percentage of cells and firing frequency were higher than the controls. Furthermore, the frequency of evoked action potentials was also higher than the controls. Co-injection of the TNF-α receptor 1 (TNFR1) inhibitor R7050 eliminated the TNF-α-induced effects, suggesting that TNF-α may directly act on RGCs to induce cell hyperexcitability through activating TNFR1. In RGCs acutely isolated from TNF-α-injected retinas, Na+ current densities were upregulated. Perfusing TNF-α in RGCs of normal rats mimicked this effect, and the activation curve of Na+ currents shifted toward hyperpolarization direction, which was mediated through p38 MAPK and STAT3 signaling pathways. Further analysis revealed that TNF-α selectively upregulated Nav1.6 subtype of Na+ currents in RGCs. Similar to observations in retinas of rats with COH, intravitreal injection of TNF-α upregulated the expression of Nav1.6 proteins in both total cell and membrane components, which was reversed by the NF-κB inhibitor BAY 11-7082. Inhibition of TNFR1 blocked TNF-α-induced RGC apoptosis. CONCLUSIONS TNF-α/TNFR1 signaling induces RGC hyperexcitability by selectively upregulating Nav1.6 Na+ channels, thus contributing to RGC apoptosis in glaucoma.
Collapse
Affiliation(s)
- Shuo Cheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Hong-Ning Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Lin-Jie Xu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Fang Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Yanying Miao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Bo Lei
- Institute of Neuroscience and Third Affiliated Hospital, Henan Provincial People’s Hospital, Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450003 China
| | - Xinghuai Sun
- Department of Ophthalmology at Eye & ENT Hospital, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031 China
| | - Zhongfeng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| |
Collapse
|
11
|
Physiological separation of Alzheimer's disease and Alzheimer's disease with significant levels of cerebrovascular symptomology and healthy controls. Med Biol Eng Comput 2021; 59:1597-1610. [PMID: 34263439 DOI: 10.1007/s11517-021-02409-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 07/04/2021] [Indexed: 01/14/2023]
Abstract
Most dementia patients with a mixed dementia (MxD) diagnosis have a mix of Alzheimer's disease (AD) and vascular dementia. Electrovestibulography (EVestG) records vestibuloacoustic afferent activity. We hypothesize EVestG recordings of AD and MxD patients are different. All patients were assessed with the Montreal cognitive assessment (MoCA) and Hachinski ischemic scale (HIS) (> 4 HIS score < 7 is representative of MxD cerebrovascular symptomology). EVestG recordings were made from 26 AD, 21 MxD and 44 healthy (control) participants. Features were derived from the EVestG recordings of the average field potential and field potential interval histogram to classify the AD, MxD and control groups. Multivariate analysis was used to test the features' significance. Using a leave-one-out cross-validated linear discriminant analysis with 3 EVestG features yielded accuracies > 80% for separating pairs of AD/MxD/control. Using the MoCA assessment and 2 EVestG features, a best accuracy of 81 to 91% depending on the classifier was obtained for the 3-way identification of AD, MxD and controls. EVestG measures provide a physiological basis for identifying AD from MxD. EVestG measures are hypothesized to be partly related to channelopathies and changes in the descending input to the vestibular periphery. Four of the five AD or MxD versus control features used had significant correlations with the MoCA. This supports assertions that the pathologic changes associated with AD impact the vestibular system and further are suggestive that the postulated physiological changes behind these features have an association with cognitive decline severity.
Collapse
|
12
|
Gelman S, Palma J, Ghavami A. Axonal Conduction Velocity in CA1 Area of Hippocampus is Reduced in Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2020; 77:1383-1388. [PMID: 32925062 DOI: 10.3233/jad-200661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The timing of action potentials arrival at synaptic terminals partially determines integration of synaptic inputs and is important for information processing in the CNS. Therefore, axonal conduction velocity (VC) is a salient parameter, influencing the timing of synaptic inputs. Even small changes in VC may disrupt information coding in networks requiring accurate timing. We recorded compound action potentials in hippocampal slices to measure VC in three mouse models of Alzheimer's disease. We report an age-dependent reduction in VC in area CA1 in two amyloid-β precursor protein transgenic mouse models, line 41 and APP/PS1, and in a tauopathy model, rTg4510.
Collapse
|
13
|
Chen WT, Lu A, Craessaerts K, Pavie B, Sala Frigerio C, Corthout N, Qian X, Laláková J, Kühnemund M, Voytyuk I, Wolfs L, Mancuso R, Salta E, Balusu S, Snellinx A, Munck S, Jurek A, Fernandez Navarro J, Saido TC, Huitinga I, Lundeberg J, Fiers M, De Strooper B. Spatial Transcriptomics and In Situ Sequencing to Study Alzheimer's Disease. Cell 2020; 182:976-991.e19. [PMID: 32702314 DOI: 10.1016/j.cell.2020.06.038] [Citation(s) in RCA: 443] [Impact Index Per Article: 110.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 04/17/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022]
Abstract
Although complex inflammatory-like alterations are observed around the amyloid plaques of Alzheimer's disease (AD), little is known about the molecular changes and cellular interactions that characterize this response. We investigate here, in an AD mouse model, the transcriptional changes occurring in tissue domains in a 100-μm diameter around amyloid plaques using spatial transcriptomics. We demonstrate early alterations in a gene co-expression network enriched for myelin and oligodendrocyte genes (OLIGs), whereas a multicellular gene co-expression network of plaque-induced genes (PIGs) involving the complement system, oxidative stress, lysosomes, and inflammation is prominent in the later phase of the disease. We confirm the majority of the observed alterations at the cellular level using in situ sequencing on mouse and human brain sections. Genome-wide spatial transcriptomics analysis provides an unprecedented approach to untangle the dysregulated cellular network in the vicinity of pathogenic hallmarks of AD and other brain diseases.
Collapse
Affiliation(s)
- Wei-Ting Chen
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Ashley Lu
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Katleen Craessaerts
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Benjamin Pavie
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium; VIB Bio Imaging Core, Gent 9052, Belgium; VIB Bio Imaging Core, Leuven 3000, Belgium
| | - Carlo Sala Frigerio
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium; UK Dementia Research Institute at University College London, London WC1E 6BT, UK
| | - Nikky Corthout
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium; VIB Bio Imaging Core, Gent 9052, Belgium; VIB Bio Imaging Core, Leuven 3000, Belgium
| | | | | | | | - Iryna Voytyuk
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Leen Wolfs
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Renzo Mancuso
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Evgenia Salta
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Sriram Balusu
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - An Snellinx
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Sebastian Munck
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium; VIB Bio Imaging Core, Gent 9052, Belgium; VIB Bio Imaging Core, Leuven 3000, Belgium
| | - Aleksandra Jurek
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Solna, Stockholm 17121, Sweden
| | - Jose Fernandez Navarro
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Solna, Stockholm 17121, Sweden
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama 351-0198, Japan
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, the Netherlands; Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1098XH, the Netherlands
| | - Joakim Lundeberg
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Solna, Stockholm 17121, Sweden
| | - Mark Fiers
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium; UK Dementia Research Institute at University College London, London WC1E 6BT, UK.
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium; UK Dementia Research Institute at University College London, London WC1E 6BT, UK.
| |
Collapse
|
14
|
Ferreira S, Pitman KA, Wang S, Summers BS, Bye N, Young KM, Cullen CL. Amyloidosis is associated with thicker myelin and increased oligodendrogenesis in the adult mouse brain. J Neurosci Res 2020; 98:1905-1932. [PMID: 32557778 PMCID: PMC7540704 DOI: 10.1002/jnr.24672] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/03/2020] [Accepted: 05/25/2020] [Indexed: 12/15/2022]
Abstract
In Alzheimer's disease, amyloid plaque formation is associated with the focal death of oligodendrocytes and soluble amyloid β impairs the survival of oligodendrocytes in vitro. However, the response of oligodendrocyte progenitor cells (OPCs) to early amyloid pathology remains unclear. To explore this, we performed a histological, electrophysiological, and behavioral characterization of transgenic mice expressing a pathological form of human amyloid precursor protein (APP), containing three single point mutations associated with the development of familial Alzheimer's disease (PDGFB‐APPSw.Ind, also known as J20 mice). PDGFB‐APPSw.Ind transgenic mice had impaired survival from weaning, were hyperactive by 2 months of age, and developed amyloid plaques by 6 months of age, however, their spatial memory remained intact over this time course. Hippocampal OPC density was normal in P60‐P180 PDGFB‐APPSw.Ind transgenic mice and, by performing whole‐cell patch‐clamp electrophysiology, we found that their membrane properties, including their response to kainate (100 µM), were largely normal. However, by P100, the response of hippocampal OPCs to GABA was elevated in PDGFB‐APPSw.Ind transgenic mice. We also found that the nodes of Ranvier were shorter, the paranodes longer, and the myelin thicker for hippocampal axons in young adult PDGFB‐APPSw.Ind transgenic mice compared with wildtype littermates. Additionally, oligodendrogenesis was normal in young adulthood, but increased in the hippocampus, entorhinal cortex, and fimbria of PDGFB‐APPSw.Ind transgenic mice as pathology developed. As the new oligodendrocytes were not associated with a change in total oligodendrocyte number, these cells are likely required for cell replacement.
Collapse
Affiliation(s)
- Solène Ferreira
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Shiwei Wang
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Benjamin S Summers
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Nicole Bye
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
15
|
Jiang R, Wu XF, Wang B, Guan RX, Lv LM, Li AP, Lei L, Ma Y, Li N, Li QF, Ma QH, Zhao J, Li S. Reduction of NgR in perforant path decreases amyloid-β peptide production and ameliorates synaptic and cognitive deficits in APP/PS1 mice. Alzheimers Res Ther 2020; 12:47. [PMID: 32331528 PMCID: PMC7181577 DOI: 10.1186/s13195-020-00616-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/07/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Amyloid beta (Aβ) which is recognized as a main feature of Alzheimer's disease (AD) has been proposed to "spread" through anatomically and functionally connected brain regions. The entorhinal cortex and perforant path are the earliest affected brain regions in AD. The perforant path is the most vulnerable circuit in the cortex with respect to both aging and AD. Previous data show that the origins and terminations of the perforant path are susceptible to amyloid deposition at the younger age in AD. Nogo receptor (NgR) plays an essential role in limiting injury-induced axonal growth and experience-dependent plasticity in the adult brain. It has been suggested that NgR is involved in AD pathological features, but the results have been conflicting and the detailed mechanism needs further investigation. In this study, the effect of NgR in the perforant path on the pathological and functional phenotype of APP/PS1 transgenic mice was studied. METHODS To genetically manipulate NgR expression, adeno-associated virus (AAV) with short hairpin (shRNA) against NgR was injected into the perforant path of APP/PS1 transgenic mice, followed by an assessment of behavioral, synaptic plasticity and neuropathological phenotypes. NgR was overexpressed or knockdown in neuroblastoma N2a cells and APPswe/HEK293 cells to investigate the interaction between NgR and amyloid precursor protein (APP). RESULTS It is shown that reduction of NgR in the perforant path rescued cognitive and synaptic deficits in APP/PS1 transgenic mice. Concurrently, Aβ production in the perforant path and levels of soluble Aβ and amyloid plaques in the hippocampus were significantly decreased. There was a positive correlation between the total APP protein level and NgR expression both in transgenic mice and in cultured cells, where the α-secretase and β-secretase cleavage products both changed with APP level in parallel. Finally, NgR might inhibit APP degradation through lysosome by Rho/Rho-associated protein kinases (ROCK) signaling pathway. CONCLUSIONS Our findings demonstrate that perforant path NgR plays an important role in regulating APP/Aβ level and cognitive functions in AD transgenic mice, which might be related to the suppression of APP degradation by NgR. Our study suggests that NgR in the perforant path could be a potential target for modulating AD progression.
Collapse
Affiliation(s)
- Rong Jiang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xue-Fei Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Bin Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Rong-Xiao Guan
- National-Local Joint Engineering Research Center for Drug Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Lang-Man Lv
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ai-Ping Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Lei Lei
- National-Local Joint Engineering Research Center for Drug Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Ye Ma
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Na Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Qi-Fa Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
16
|
Thetiot M, Freeman SA, Roux T, Dubessy AL, Aigrot MS, Rappeneau Q, Lejeune FX, Tailleur J, Sol-Foulon N, Lubetzki C, Desmazieres A. An alternative mechanism of early nodal clustering and myelination onset in GABAergic neurons of the central nervous system. Glia 2020; 68:1891-1909. [PMID: 32119167 DOI: 10.1002/glia.23812] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/12/2020] [Accepted: 02/19/2020] [Indexed: 01/06/2023]
Abstract
In vertebrates, fast saltatory conduction along myelinated axons relies on the node of Ranvier. How nodes assemble on CNS neurons is not yet fully understood. We previously described that node-like clusters can form prior to myelin deposition in hippocampal GABAergic neurons and are associated with increased conduction velocity. Here, we used a live imaging approach to characterize the intrinsic mechanisms underlying the assembly of these clusters prior to myelination. We first demonstrated that their components can partially preassemble prior to membrane targeting and determined the molecular motors involved in their trafficking. We then demonstrated the key role of the protein β2Nav for node-like clustering initiation. We further assessed the fate of these clusters when myelination proceeds. Our results shed light on the intrinsic mechanisms involved in node-like clustering prior to myelination and unravel a potential role of these clusters in node of Ranvier formation and in guiding myelination onset.
Collapse
Affiliation(s)
- Melina Thetiot
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière, ICM-GH Pitié-Salpêtrière, Paris, France
| | - Sean A Freeman
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière, ICM-GH Pitié-Salpêtrière, Paris, France.,Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Thomas Roux
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière, ICM-GH Pitié-Salpêtrière, Paris, France.,Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Anne-Laure Dubessy
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière, ICM-GH Pitié-Salpêtrière, Paris, France.,Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Marie-Stéphane Aigrot
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière, ICM-GH Pitié-Salpêtrière, Paris, France
| | - Quentin Rappeneau
- Sorbonne Université, UPMC Paris 06, Inserm, CNRS, Institut de la Vision, Paris, France
| | - François-Xavier Lejeune
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière, ICM-GH Pitié-Salpêtrière, Paris, France
| | - Julien Tailleur
- Université Paris Diderot, Sorbonne Paris Cité, MSC, UMR 7057 CNRS, Paris, France
| | - Nathalie Sol-Foulon
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière, ICM-GH Pitié-Salpêtrière, Paris, France
| | - Catherine Lubetzki
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière, ICM-GH Pitié-Salpêtrière, Paris, France.,Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Anne Desmazieres
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle épinière, ICM-GH Pitié-Salpêtrière, Paris, France
| |
Collapse
|
17
|
Truong PH, Ciccotosto GD, Merson TD, Spoerri L, Chuei MJ, Ayers M, Xing YL, Emery B, Cappai R. Amyloid precursor protein and amyloid precursor-like protein 2 have distinct roles in modulating myelination, demyelination, and remyelination of axons. Glia 2018; 67:525-538. [DOI: 10.1002/glia.23561] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Phan H. Truong
- Department of Pathology; The University of Melbourne; Melbourne Victoria Australia
- The Bio21 Molecular Science and Biotechnology Institute; The University of Melbourne; Melbourne Victoria Australia
- Department of Pharmacology and Therapeutics; The University of Melbourne; Melbourne Victoria Australia
| | - Giuseppe D. Ciccotosto
- Department of Pathology; The University of Melbourne; Melbourne Victoria Australia
- The Bio21 Molecular Science and Biotechnology Institute; The University of Melbourne; Melbourne Victoria Australia
- Department of Pharmacology and Therapeutics; The University of Melbourne; Melbourne Victoria Australia
| | - Tobias D. Merson
- The Florey Institute of Neuroscience and Mental Health; The University of Melbourne; Melbourne Victoria Australia
| | - Loredana Spoerri
- Department of Pathology; The University of Melbourne; Melbourne Victoria Australia
- The Bio21 Molecular Science and Biotechnology Institute; The University of Melbourne; Melbourne Victoria Australia
| | - Mun Joo Chuei
- Department of Pathology; The University of Melbourne; Melbourne Victoria Australia
- The Bio21 Molecular Science and Biotechnology Institute; The University of Melbourne; Melbourne Victoria Australia
| | - Margaret Ayers
- Department of Pathology; The University of Melbourne; Melbourne Victoria Australia
| | - Yao Lulu Xing
- The Florey Institute of Neuroscience and Mental Health; The University of Melbourne; Melbourne Victoria Australia
| | - Ben Emery
- The Florey Institute of Neuroscience and Mental Health; The University of Melbourne; Melbourne Victoria Australia
- Department of Anatomy and Neuroscience; The University of Melbourne; Melbourne Victoria Australia
| | - Roberto Cappai
- Department of Pathology; The University of Melbourne; Melbourne Victoria Australia
- The Bio21 Molecular Science and Biotechnology Institute; The University of Melbourne; Melbourne Victoria Australia
- Department of Pharmacology and Therapeutics; The University of Melbourne; Melbourne Victoria Australia
| |
Collapse
|
18
|
Liu CB, Wang R, Yi YF, Gao Z, Chen YZ. Lycopene mitigates β-amyloid induced inflammatory response and inhibits NF-κB signaling at the choroid plexus in early stages of Alzheimer's disease rats. J Nutr Biochem 2017; 53:66-71. [PMID: 29195132 DOI: 10.1016/j.jnutbio.2017.10.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/28/2017] [Accepted: 10/24/2017] [Indexed: 11/17/2022]
Abstract
The choroid plexus is able to modulate the cognitive function, through changes in the neuroinflammatory response and in brain immune surveillance. However, whether lycopene is involved in inflammatory responses at the choroid plexus in the early stages of Alzheimer's disease, and its molecular underpinnings are elusive. In this rat study, lycopene was used to investigate its protective effects on inflammation caused by β-amyloid. We characterized the learning and memory abilities, cytokine profiles of circulating TNF-α, IL-1β and IL-6β in the serum and the expressions of Toll like receptor 4 and nuclear factor-κB p65 mRNA and protein at the choroid plexus. The results showed that functional deficits of learning and memory in lycopene treatment groups were significantly improved compared to the control group without lycopene treatment in water maze test. The levels of serum TNF-α, IL-1β and IL-6β were significantly increased, and the expressions of TLR4 and NF-κB p65 mRNA and protein at the choroid plexus were up-regulated, indicating inflammation response was initiated following administration of Aβ1-42. After intragastric pretreatment with lycopene, inflammatory cytokines were significantly reduced and lycopene also reversed the Aβ1-42 induced up-regulation of TLR4 and NF-κB p65 mRNA and protein expressions at the choroid plexus. These results provided a novel evidence that lycopene significantly improved cognitive deficits and were accompanied by the attenuation of inflammatory injury via blocking the activation of NF-κB p65 and TLR4 expressions and production of cytokines, thereby endorsing its usefulness for diminishing β-amyloid deposition in the hippocampus tissues.
Collapse
Affiliation(s)
- Chong-Bin Liu
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China; Department of Physiology, Basic Medical Science College, Wenzhou Medical University, 325035, Wenzhou, People's Republic of China.
| | - Rui Wang
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China
| | - Yan-Feng Yi
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China
| | - Zhen Gao
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China
| | - Yi-Zhu Chen
- Central Blood Station of Huzhou City, 313020, Huzhou, People's Republic of China.
| |
Collapse
|
19
|
Arancibia-Cárcamo IL, Ford MC, Cossell L, Ishida K, Tohyama K, Attwell D. Node of Ranvier length as a potential regulator of myelinated axon conduction speed. eLife 2017; 6. [PMID: 28130923 PMCID: PMC5313058 DOI: 10.7554/elife.23329] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/24/2017] [Indexed: 12/30/2022] Open
Abstract
Myelination speeds conduction of the nerve impulse, enhancing cognitive power. Changes of white matter structure contribute to learning, and are often assumed to reflect an altered number of myelin wraps. We now show that, in rat optic nerve and cerebral cortical axons, the node of Ranvier length varies over a 4.4-fold and 8.7-fold range respectively and that variation of the node length is much less along axons than between axons. Modelling predicts that these node length differences will alter conduction speed by ~20%, similar to the changes produced by altering the number of myelin wraps or the internode length. For a given change of conduction speed, the membrane area change needed at the node is >270-fold less than that needed in the myelin sheath. Thus, axon-specific adjustment of node of Ranvier length is potentially an energy-efficient and rapid mechanism for tuning the arrival time of information in the CNS. DOI:http://dx.doi.org/10.7554/eLife.23329.001 Information is transmitted around the nervous system as electrical signals passing along nerve cells. A fatty substance called myelin, which is wrapped around the nerve cells, increases the speed with which the signals travel along the nerve cells. This allows us to think and move faster than we would otherwise be able to do. The electrical signals start at small “nodes” between areas of myelin wrapping. Originally it was thought that we learn things mainly as a result of changes in the strength of connections between nerve cells, but recently it has been proposed that changes in myelin wrapping could also contribute to learning. Arancibia-Cárcamo, Ford, Cossell et al. investigated how much node structure varies in rat nerve cells, and whether differences in the length of nodes can fine-tune the activity of the nervous system. The experiments show that rat nerve cells do indeed have nodes with a range of different lengths. Calculations show that this could result in electrical signals moving at different speeds through different nerve cells. These findings raise the possibility that nerve cells actively alter the length of their nodes in order to alter their signal speed. The next step is to try to show experimentally that this happens during learning in animals. DOI:http://dx.doi.org/10.7554/eLife.23329.002
Collapse
Affiliation(s)
- I Lorena Arancibia-Cárcamo
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Marc C Ford
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Lee Cossell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Kinji Ishida
- The Center for Electron Microscopy and Bio-Imaging Research, Iwate Medical University, Morioka, Japan
| | - Koujiro Tohyama
- The Center for Electron Microscopy and Bio-Imaging Research, Iwate Medical University, Morioka, Japan.,Department of Physiology, School of Dentistry, Iwate Medical University, Morioka, Japan
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
20
|
Li S, Wang X, Ma QH, Yang WL, Zhang XG, Dawe GS, Xiao ZC. Amyloid precursor protein modulates Nav1.6 sodium channel currents through a Go-coupled JNK pathway. Sci Rep 2016; 6:39320. [PMID: 28008944 PMCID: PMC5180232 DOI: 10.1038/srep39320] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 11/17/2016] [Indexed: 02/08/2023] Open
Abstract
Amyloid precursor protein (APP), commonly associated with Alzheimer's disease, also marks axonal degeneration. In the recent studies, we demonstrated that APP aggregated at nodes of Ranvier (NORs) in myelinated central nervous system (CNS) axons and interacted with Nav1.6. However, the physiological function of APP remains unknown. In this study, we described reduced sodium current densities in APP knockout hippocampal neurons. Coexpression of APP or its intracellular domains containing a VTPEER motif with Nav1.6 sodium channels in Xenopus oocytes resulted in an increase in peak sodium currents, which was enhanced by constitutively active Go mutant and blocked by a dominant negative mutant. JNK and CDK5 inhibitor attenuated increases in Nav1.6 sodium currents induced by overexpression of APP. Nav1.6 sodium currents were increased by APPT668E (mutant Thr to Glu) and decreased by T668A (mutant Thr to ALa) mutant, respectively. The cell surface expression of Nav1.6 sodium channels in the white matter of spinal cord and the spinal conduction velocity is decreased in APP, p35 and JNK3 knockout mice. Therefore, APP modulates Nav1.6 sodium channels through a Go-coupled JNK pathway, which is dependent on phosphorylation of APP at Thr668.
Collapse
Affiliation(s)
- Shao Li
- Department of Physiology, Dalian Medical University, Dalian 116044, China
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 10 Medical Drive 117597, Singapore
| | - Xi Wang
- Department of Physiology, Dalian Medical University, Dalian 116044, China
- The Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical College, Kunming 650031, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Wu-lin Yang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 10 Medical Drive 117597, Singapore
| | - Xiao-Gang Zhang
- Department of Physiology, Dalian Medical University, Dalian 116044, China
| | - Gavin S. Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 10 Medical Drive 117597, Singapore
- Neurobiology and Ageing Programme, Life Sciences Institute, Centre for Life Sciences, National University of Singapore, 28 Medical Drive 117456, Singapore
- Singapore Institute for Neurotechnology (SINAPSE), Centre for Life Sciences, National University of Singapore, 28 Medical Drive 117456, Singapore
| | - Zhi-Cheng Xiao
- The Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical College, Kunming 650031, China
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| |
Collapse
|
21
|
Ayyadevara S, Balasubramaniam M, Parcon PA, Barger SW, Griffin WST, Alla R, Tackett AJ, Mackintosh SG, Petricoin E, Zhou W, Shmookler Reis RJ. Proteins that mediate protein aggregation and cytotoxicity distinguish Alzheimer's hippocampus from normal controls. Aging Cell 2016; 15:924-39. [PMID: 27448508 PMCID: PMC5013017 DOI: 10.1111/acel.12501] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2016] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative diseases are distinguished by characteristic protein aggregates initiated by disease‐specific ‘seed’ proteins; however, roles of other co‐aggregated proteins remain largely unexplored. Compact hippocampal aggregates were purified from Alzheimer's and control‐subject pools using magnetic‐bead immunoaffinity pulldowns. Their components were fractionated by electrophoretic mobility and analyzed by high‐resolution proteomics. Although total detergent‐insoluble aggregates from Alzheimer's and controls had similar protein content, within the fractions isolated by tau or Aβ1–42 pulldown, the protein constituents of Alzheimer‐derived aggregates were more abundant, diverse, and post‐translationally modified than those from controls. Tau‐ and Aβ‐containing aggregates were distinguished by multiple components, and yet shared >90% of their protein constituents, implying similar accretion mechanisms. Alzheimer‐specific protein enrichment in tau‐containing aggregates was corroborated for individuals by three analyses. Five proteins inferred to co‐aggregate with tau were confirmed by precise in situ methods, including proximity ligation amplification that requires co‐localization within 40 nm. Nematode orthologs of 21 proteins, which showed Alzheimer‐specific enrichment in tau‐containing aggregates, were assessed for aggregation‐promoting roles in C. elegans by RNA‐interference ‘knockdown’. Fifteen knockdowns (71%) rescued paralysis of worms expressing muscle Aβ, and 12 (57%) rescued chemotaxis disrupted by neuronal Aβ expression. Proteins identified in compact human aggregates, bound by antibody to total tau, were thus shown to play causal roles in aggregation based on nematode models triggered by Aβ1–42. These observations imply shared mechanisms driving both types of aggregation, and/or aggregate‐mediated cross‐talk between tau and Aβ. Knowledge of protein components that promote protein accrual in diverse aggregate types implicates common mechanisms and identifies novel targets for drug intervention.
Collapse
Affiliation(s)
- Srinivas Ayyadevara
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205 USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Meenakshisundaram Balasubramaniam
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
- BioInformatics Program University of Arkansas for Medical Sciences and University of Arkansas at Little Rock Little Rock AR 72205 USA
| | - Paul A. Parcon
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Steven W. Barger
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205 USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - W. Sue T. Griffin
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205 USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Ramani Alla
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205 USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Alan J. Tackett
- Department of Biochemistry & Molecular Biology University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Samuel G. Mackintosh
- Department of Biochemistry & Molecular Biology University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Emanuel Petricoin
- Center for Applied Proteomics and Molecular Medicine George Mason University Manassas VA 20110 USA
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine George Mason University Manassas VA 20110 USA
| | - Robert J. Shmookler Reis
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205 USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
- Department of Biochemistry & Molecular Biology University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| |
Collapse
|
22
|
Echeverria V, Yarkov A, Aliev G. Positive modulators of the α7 nicotinic receptor against neuroinflammation and cognitive impairment in Alzheimer's disease. Prog Neurobiol 2016; 144:142-57. [DOI: 10.1016/j.pneurobio.2016.01.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 11/07/2015] [Accepted: 01/06/2016] [Indexed: 01/08/2023]
|
23
|
Matías-Guiu JA, Oreja-Guevara C, Cabrera-Martín MN, Moreno-Ramos T, Carreras JL, Matías-Guiu J. Amyloid Proteins and Their Role in Multiple Sclerosis. Considerations in the Use of Amyloid-PET Imaging. Front Neurol 2016; 7:53. [PMID: 27065425 PMCID: PMC4814935 DOI: 10.3389/fneur.2016.00053] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 03/22/2016] [Indexed: 02/06/2023] Open
Abstract
Thioflavin T derivatives are used in positron-emission tomography (PET) studies to detect amyloid protein deposits in patients with Alzheimer disease. These tracers bind extensively to white matter, which suggests that they may be useful in studies of multiple sclerosis (MS), and that proteins resulting from proteolytic processing of the amyloid precursor protein (APP) may contribute to MS. This article reviews data from both clinical and preclinical studies addressing the role of these proteins, whether they are detected in CSF studies or using PET imaging. APP is widely expressed in demyelinated axons and may have a protective effect in MS and in experimental allergic encephalomyelitis in animals. Several mechanisms associated with this increased expression may affect the degree of remyelination in MS. Amyloid-PET imaging may help determine the degree of demyelination and provide information on the molecular changes linked to APP proteolytic processing experienced by patients with MS.
Collapse
Affiliation(s)
- Jordi A Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Celia Oreja-Guevara
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - María Nieves Cabrera-Martín
- Department of Nuclear Medicine, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Teresa Moreno-Ramos
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - José Luis Carreras
- Department of Nuclear Medicine, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Jorge Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| |
Collapse
|
24
|
Wang X, Zhang XG, Zhou TT, Li N, Jang CY, Xiao ZC, Ma QH, Li S. Elevated Neuronal Excitability Due to Modulation of the Voltage-Gated Sodium Channel Nav1.6 by Aβ1-42. Front Neurosci 2016; 10:94. [PMID: 27013956 PMCID: PMC4783403 DOI: 10.3389/fnins.2016.00094] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/24/2016] [Indexed: 12/13/2022] Open
Abstract
Aberrant increases in neuronal network excitability may contribute to the cognitive deficits in Alzheimer's disease (AD). However, the mechanisms underlying hyperexcitability are not fully understood. Such overexcitation of neuronal networks has been detected in the brains of APP/PS1 mice. In the present study, using current-clamp recording techniques, we observed that 12 days in vitro (DIV) primary cultured pyramidal neurons from P0 APP/PS1 mice exhibited a more prominent action potential burst and a lower threshold than WT littermates. Moreover, after treatment with Aβ1−42 peptide, 12 DIV primary cultured neurons showed similar changes, to a greater degree than in controls. Voltage-clamp recordings revealed that the voltage-dependent sodium current density of neurons incubated with Aβ1−42 was significantly increased, without change in the voltage-dependent sodium channel kinetic characteristics. Immunohistochemistry and western blot results showed that, after treatment with Aβ1−42, expressions of Nav and Nav1.6 subtype increased in cultured neurons or APP/PS1 brains compared to control groups. The intrinsic neuronal hyperexcitability of APP/PS1 mice might thus be due to an increased expression of voltage-dependent sodium channels induced by Aβ1−42. These results may illuminate the mechanism of aberrant neuronal networks in AD.
Collapse
Affiliation(s)
- Xi Wang
- Department of Physiology, Dalian Medical University Dalian, China
| | - Xiao-Gang Zhang
- Department of Physiology, Dalian Medical University Dalian, China
| | - Ting-Ting Zhou
- Department of Neurology, the First Affiliated Hospital of Dalian Medical University Dalian, China
| | - Na Li
- Department of Physiology, Dalian Medical University Dalian, China
| | - Chun-Yan Jang
- Department of Physiology, Dalian Medical University Dalian, China
| | - Zhi-Cheng Xiao
- The Key Laboratory of Stem Cell and Regenerative Medicine, Kunming Medical College, Institute of Molecular and Clinical Medicine Kunming, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University Suzhou, China
| | - Shao Li
- Department of Physiology, Dalian Medical University Dalian, China
| |
Collapse
|
25
|
Liu C, Tan FCK, Xiao ZC, Dawe GS. Amyloid precursor protein enhances Nav1.6 sodium channel cell surface expression. J Biol Chem 2015; 290:12048-57. [PMID: 25767117 DOI: 10.1074/jbc.m114.617092] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Indexed: 12/19/2022] Open
Abstract
Amyloid precursor protein (APP) is commonly associated with Alzheimer disease, but its physiological function remains unknown. Nav1.6 is a key determinant of neuronal excitability in vivo. Because mouse models of gain of function and loss of function of APP and Nav1.6 share some similar phenotypes, we hypothesized that APP might be a candidate molecule for sodium channel modulation. Here we report that APP colocalized and interacted with Nav1.6 in mouse cortical neurons. Knocking down APP decreased Nav1.6 sodium channel currents and cell surface expression. APP-induced increases in Nav1.6 cell surface expression were Go protein-dependent, enhanced by a constitutively active Go protein mutant, and blocked by a dominant negative Go protein mutant. APP also regulated JNK activity in a Go protein-dependent manner. JNK inhibition attenuated increases in cell surface expression of Nav1.6 sodium channels induced by overexpression of APP. JNK, in turn, phosphorylated APP. Nav1.6 sodium channel surface expression was increased by T668E and decreased by T668A, mutations of APP695 mimicking and preventing Thr-668 phosphorylation, respectively. Phosphorylation of APP695 at Thr-668 enhanced its interaction with Nav1.6. Therefore, we show that APP enhances Nav1.6 sodium channel cell surface expression through a Go-coupled JNK pathway.
Collapse
Affiliation(s)
- Chao Liu
- From the Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, the Neurobiology and Ageing Programme, Life Sciences Institute and Singapore Institute for Neurotechnology (SINAPSE), Centre for Life Sciences, National University of Singapore, 28 Medical Drive, Singapore 117456
| | - Francis Chee Kuan Tan
- From the Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, the Neurobiology and Ageing Programme, Life Sciences Institute and Singapore Institute for Neurotechnology (SINAPSE), Centre for Life Sciences, National University of Singapore, 28 Medical Drive, Singapore 117456
| | - Zhi-Cheng Xiao
- the Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical College, Kunming 650031, China, and the Shunxi-Monash Immune Regeneration and Neuroscience Laboratories, Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Gavin S Dawe
- From the Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, the Neurobiology and Ageing Programme, Life Sciences Institute and Singapore Institute for Neurotechnology (SINAPSE), Centre for Life Sciences, National University of Singapore, 28 Medical Drive, Singapore 117456,
| |
Collapse
|
26
|
Xu CJ, Wang JL, Jin WL. The Neural Stem Cell Microenvironment: Focusing on Axon Guidance Molecules and Myelin-Associated Factors. J Mol Neurosci 2015; 56:887-897. [PMID: 25757451 DOI: 10.1007/s12031-015-0538-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/27/2015] [Indexed: 12/20/2022]
Abstract
Neural stem cells (NSCs) could produce various cell phenotypes in the subventricular zone (SVZ) and dentate gyrus of the hippocampus in the central nervous system (CNS), where neurogenesis has been determined to occur. The extracellular microenvironment also influences the behaviors of NSCs during development and at CNS injury sites. Our previous study indicates that myelin, a component of the CNS, could regulate the differentiation of NSCs in vitro. Recent reports have implicated three myelin-derived inhibitors, NogoA, myelin-associated glycoprotein (MAG), and oligodendrocyte-myelin glycoprotein (OMgp), as well as several axon guidance molecules as regulators of NSC survival, proliferation, migration, and differentiation. However, the molecular mechanisms underlying the behavior of NSCs are not fully understood. In this study, we summarize the current literature on the effects of different extrinsic factors on NSCs and discuss possible mechanisms, as well as future possible clinical applications.
Collapse
Affiliation(s)
- Chao-Jin Xu
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, University town, Cha Shan, Zhejiang, 325035, China.
| | - Jun-Ling Wang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China. .,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, 200240, China.
| |
Collapse
|