1
|
Kopalli SR, Behl T, Baldaniya L, Ballal S, Joshi KK, Arya R, Chaturvedi B, Chauhan AS, Verma R, Patel M, Jain SK, Wal A, Gulati M, Koppula S. Neuroadaptation in neurodegenerative diseases: compensatory mechanisms and therapeutic approaches. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111375. [PMID: 40280271 DOI: 10.1016/j.pnpbp.2025.111375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Progressive neuronal loss is a hallmark of neurodegenerative diseases including Alzheimer's, Parkinson's, Huntington's, and Amyotrophic Lateral Sclerosis (ALS), which cause cognitive and motor impairment. Delaying the onset and course of symptoms is largely dependent on neuroadaptation, the brain's ability to restructure in response to damage. The molecular, cellular, and systemic processes that underlie neuroadaptation are examined in this study. These mechanisms include gliosis, neurogenesis, synaptic plasticity, and changes in neurotrophic factors. Axonal sprouting, dendritic remodelling, and compensatory alterations in neurotransmitter systems are important adaptations observed in NDDs; nevertheless, these processes may shift to maladaptive plasticity, which would aid in the advancement of the illness. Amyloid and tau pathology-induced synaptic alterations in Alzheimer's disease emphasize compensatory network reconfiguration. Dopamine depletion causes a major remodelling of the basal ganglia in Parkinson's disease, and non-dopaminergic systems compensate. Both ALS and Huntington's disease rely on motor circuit rearrangement and transcriptional dysregulation to slow down functional deterioration. Neuroadaptation is, however, constrained by oxidative stress, compromised autophagy, and neuroinflammation, particularly in elderly populations. The goal of emerging therapy strategies is to improve neuroadaptation by pharmacologically modifying neurotrophic factors, neuroinflammation, and synaptic plasticity. Neurostimulation, cognitive training, and physical rehabilitation are instances of non-pharmacological therapies that support neuroplasticity. Restoring compensating systems may be possible with the use of stem cell techniques and new gene treatments. The goal of future research is to combine biomarkers and individualized medicines to maximize neuroadaptive responses and decrease the course of illness. In order to reduce neurodegeneration and enhance patient outcomes, this review highlights the dual function of neuroadaptation in NDDs and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Punjab-140306, India
| | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Bhumi Chaturvedi
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Rakesh Verma
- Department of Pharmacology, Institute of Medical Science, BHU, Varanasi, India
| | - Minesh Patel
- Department of Pharmacology & Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Dhanap, Gandhinagar, Gujarat, India
| | - Sanmati Kumar Jain
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Koni, Bilaspur, India, 495009
| | - Ankita Wal
- Pranveer Singh Institute of Technology, Pharmacy, NH-19, Bhauti Road, Kanpur, UP, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
2
|
Zhang Z, Zhang S, Liu S, He Y, Wang A. Fuzhisan ameliorates cognitive ability in Alzheimer's disease by p62 and related autophagy regulatory pathways. Brain Res 2025; 1849:149436. [PMID: 39736370 DOI: 10.1016/j.brainres.2024.149436] [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: 12/11/2023] [Revised: 08/29/2024] [Accepted: 12/27/2024] [Indexed: 01/01/2025]
Abstract
BACKGROUND Maintaining autophagic homeostasis has been proved to play an important role in Alzheimer's disease. OBJECT The aim of this study was to investigate the effect of Fuzhisan(FZS) on autophagic function in Alzheimer's disease and to elucidate its potential mechanism through the P62 regulatory pathways. METHODS FZS was extracted by water extraction-rotary evaporation method. The novel object recognition test, morris water maze test and Y maze test were used to observe the cognitive and memory ability of APP/PS1 mice. The effects of FZS on the ultrastructure of mice hippocampus were examined by transmission electron microscopy. Molecular level changes were also further detected, including Aβ deposition, tau hyperphosphorylation, SOD, CAT and autophagy related proteins (p62, Nrf2, keap1, mTOR, LC3II/I, Beclin1, Atgs). RESULTS FZS could alleviate memory and cognitive impairment in APP/PS1 mice, increase the autophagic vesicles and organelle abundance in hippocampus. FZS also reduced the levels of Aβ and tau hyperphosphorylation in the hippocampus of model mice, upregulated the levels of SOD, CAT and autophagy related proteins (Nrf2, LC3II/LC3I, Beclin1, Atg7 and Atg12) as well as downregulated the expression of P62, keap1 and p-mTOR/mTOR proteins. Co-Ip results showed that FZS elevated the levels of p62/LC3 and P62-keap1-Nrf2 complex, but decreased the P62 and keap1 association. CONCLUSION Our findings indicate that FZS may affect autophagy function and oxidative stress by regulating P62 and related pathways to promote the clearance of Aβ and phosphorylated tau, thereby improving the cognitive ability of AD, which provided a novel perspective for exploring the potential mechanism of FZS upon AD.
Collapse
Affiliation(s)
- Zhaoxu Zhang
- Department of Neurology, Peking University People's Hospital, Beijing 100044, PR China
| | - Shuangmei Zhang
- Department of Pain Rehabilitation Rehabilitation, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, PR China
| | - Shen Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 251412, PR China; Department of Neurology of TCM, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Yang He
- Department of Neurology, Peking University People's Hospital, Beijing 100044, PR China
| | - Anrong Wang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan 250014, PR China.
| |
Collapse
|
3
|
Chen L, Zhang K, Liu J, Li X, Liu Y, Ma H, Yang J, Li J, Chen L, Hsu C, Zeng J, Xie X, Wang Q. The role of the microbiota-gut-brain axis in methamphetamine-induced neurotoxicity: Disruption of microbial composition and short-chain fatty acid metabolism. Acta Pharm Sin B 2024; 14:4832-4857. [PMID: 39664442 PMCID: PMC11628825 DOI: 10.1016/j.apsb.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/08/2024] [Accepted: 07/24/2024] [Indexed: 12/13/2024] Open
Abstract
Methamphetamine (METH) abuse is associated with significant neurotoxicity, high addiction potential, and behavioral abnormalities. Recent studies have identified a connection between the gut microbiota and METH-induced neurotoxicity and behavioral disorders. However, the underlying causal mechanisms linking the gut microbiota to METH pathophysiology remain largely unexplored. In this study, we employed fecal microbiota transplantation (FMT) and antibiotic (Abx) intervention to manipulate the gut microbiota in mice administered METH. Furthermore, we supplemented METH-treated mice with short-chain fatty acids (SCFAs) and pioglitazone (Pio) to determine the protective effects on gut microbiota metabolism. Finally, we assessed the underlying mechanisms of the gut-brain neural circuit in vagotomized mice. Our data provide compelling evidence that modulation of the gut microbiome through FMT or microbiome knockdown by Abx plays a crucial role in METH-induced neurotoxicity, behavioral disorders, gut microbiota disturbances, and intestinal barrier impairment. Furthermore, our findings highlight a novel prevention strategy for mitigating the risks to both the nervous and intestinal systems caused by METH, which involves supplementation with SCFAs or Pio.
Collapse
Affiliation(s)
- Lijian Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Kaikai Zhang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jiali Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiuwen Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yi Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Hongsheng Ma
- Shunde Police in Foshan City, Foshan 528300, China
| | - Jianzheng Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jiahao Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Long Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Clare Hsu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jiahao Zeng
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiaoli Xie
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), Guangzhou 510515, China
| | - Qi Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
4
|
Epp R, Glück C, Binder NF, El Amki M, Weber B, Wegener S, Jenny P, Schmid F. The role of leptomeningeal collaterals in redistributing blood flow during stroke. PLoS Comput Biol 2023; 19:e1011496. [PMID: 37871109 PMCID: PMC10621965 DOI: 10.1371/journal.pcbi.1011496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 11/02/2023] [Accepted: 09/03/2023] [Indexed: 10/25/2023] Open
Abstract
Leptomeningeal collaterals (LMCs) connect the main cerebral arteries and provide alternative pathways for blood flow during ischaemic stroke. This is beneficial for reducing infarct size and reperfusion success after treatment. However, a better understanding of how LMCs affect blood flow distribution is indispensable to improve therapeutic strategies. Here, we present a novel in silico approach that incorporates case-specific in vivo data into a computational model to simulate blood flow in large semi-realistic microvascular networks from two different mouse strains, characterised by having many and almost no LMCs between middle and anterior cerebral artery (MCA, ACA) territories. This framework is unique because our simulations are directly aligned with in vivo data. Moreover, it allows us to analyse perfusion characteristics quantitatively across all vessel types and for networks with no, few and many LMCs. We show that the occlusion of the MCA directly caused a redistribution of blood that was characterised by increased flow in LMCs. Interestingly, the improved perfusion of MCA-sided microvessels after dilating LMCs came at the cost of a reduced blood supply in other brain areas. This effect was enhanced in regions close to the watershed line and when the number of LMCs was increased. Additional dilations of surface and penetrating arteries after stroke improved perfusion across the entire vasculature and partially recovered flow in the obstructed region, especially in networks with many LMCs, which further underlines the role of LMCs during stroke.
Collapse
Affiliation(s)
- Robert Epp
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Nadine Felizitas Binder
- Deptartment of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Mohamad El Amki
- Deptartment of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Susanne Wegener
- Deptartment of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Patrick Jenny
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Franca Schmid
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Abdelmoaty MM, Yeapuri P, Machhi J, Lu Y, Namminga KL, Kadry R, Lu E, Bhattarai S, Mosley RL, Gendelman HE. Immune senescence in aged APP/PS1 mice. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:317-330. [PMID: 38023614 PMCID: PMC10659760 DOI: 10.1515/nipt-2023-0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 12/01/2023]
Abstract
Objectives To evaluate the linkage between age and deficits in innate and adaptive immunity which heralds both Alzheimer's disease (AD) onset and progression. The pathobiological events which underlie and tie these outcomes remain not fully understood. Methods To investigate age-dependent immunity in AD, we evaluated innate and adaptive immunity in coordinate studies of regulatory T cell (Treg) function, T cell frequencies, and microglial integrity. These were assessed in blood, peripheral lymphoid tissues, and the hippocampus of transgenic (Tg) amyloid precursor protein/presenilin 1 (APP/PS1) against non-Tg mice. Additionally, immune arrays of hippocampal tissue were performed at 4, 6, 12, and 20 months of age. Results APP/PS1 mice showed progressive impairment of Treg immunosuppressive function with age. There was partial restoration of Treg function in 20-month-old mice. Ingenuity pathway analyses of hippocampal tissues were enriched in inflammatory, oxidative, and cellular activation pathways that paralleled advancing age and AD-pathobiology. Operative genes in those pathways included, but were not limited to triggering receptor on myeloid cells 1 (TREM1), T helper type 1 (Th1), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways. Interleukin-17 (IL-17), nitric oxide, acute phase, and T cell receptor signaling pathways were also perturbed. Significant inflammation was observed at 6- and 12-months. However, at 20-months, age associated partial restoration of Treg function reduced inflammatory phenotype. Conclusions Impaired Treg function, inflammation and oxidative stress were associated with AD pathology. Age associated partial restoration of Treg function in old mice reduced the hippocampal inflammatory phenotype. Restoring Treg suppressive function can be a therapeutic modality for AD.
Collapse
Affiliation(s)
- Mai M. Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Krista L. Namminga
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rana Kadry
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eugene Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rodney Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
6
|
Blinowska KJ, Kamiński M, Macrez N, Marighetto A, Meyrand P, Bem T. Effect of learning on slow gamma propagation between hippocampus and cortex in the wild-type and AD mice. Sci Rep 2022; 12:22343. [PMID: 36572725 PMCID: PMC9792563 DOI: 10.1038/s41598-022-26754-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Slow gamma oscillations (20-50 Hz) have been suggested to coordinate information transfer between brain structures involved in memory formation. Whereas the involvement of slow gamma in memory processing was studied by means of correlation between the gamma power and the occurrence of a given event (sharp wave ripples (SWRs), cortical transients), our approach consists of the analysis of the transmission of slow gamma itself. We use the method based on Granger causality principle-direct Directed Transfer Function, which allows to determine directed propagation of brain activity, including bidirectional flows. Four cortical sites along with CA1 ipsi- and contralateral were recorded in behaving wild-type and APP/PS1 mice before and after learning session of a spatial memory task. During slow wave sleep propagation of slow gamma was bidirectional, forming multiple loops of interaction which involved both CA1 and some of cortical sites. In episodes coincident with SWRs the number and strength of connectivity pathways increased in both groups compared to episodes without SWRs. The effect of learning was expressed only in APP/PS1 mice and consisted in strengthening of the slow gamma transmission from hippocampus to cortex as well as between both CA1 which may serve more efficient transmission of information from impaired CA1.
Collapse
Affiliation(s)
- Katarzyna J. Blinowska
- grid.413454.30000 0001 1958 0162Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland ,grid.12847.380000 0004 1937 1290Department of Biomedical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Maciej Kamiński
- grid.12847.380000 0004 1937 1290Department of Biomedical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Nathalie Macrez
- grid.412041.20000 0001 2106 639XUniversity Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000 Bordeaux, France
| | - Aline Marighetto
- grid.412041.20000 0001 2106 639XUniversity Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000 Bordeaux, France
| | - Pierre Meyrand
- grid.412041.20000 0001 2106 639XUniversity Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000 Bordeaux, France
| | - Tiaza Bem
- grid.413454.30000 0001 1958 0162Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
7
|
Liu W, Li J, Li L, Zhang Y, Yang M, Liang S, Li L, Dai Y, Chen L, Jia W, He X, Lin H, Tao J. Enhanced Medial Prefrontal Cortex and Hippocampal Activity Improves Memory Generalization in APP/PS1 Mice: A Multimodal Animal MRI Study. Front Cell Neurosci 2022; 16:848967. [PMID: 35386301 PMCID: PMC8977524 DOI: 10.3389/fncel.2022.848967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/08/2022] [Indexed: 02/03/2023] Open
Abstract
Memory generalization allows individuals to extend previously learned movement patterns to similar environments, contributing to cognitive flexibility. In Alzheimer’s disease (AD), the disturbance of generalization is responsible for the deficits of episodic memory, causing patients with AD to forget or misplace things, even lose track of the way home. Cognitive training can effectively improve the cognition of patients with AD through changing thinking mode and memory flexibility. In this study, a T-shaped maze was utilized to simulate cognitive training in APP/PS1 mice to elucidate the potential mechanisms of beneficial effects after cognitive training. We found that cognitive training conducted by a T-shaped maze for 4 weeks can improve the memory generalization ability of APP/PS1 mice. The results of functional magnetic resonance imaging (fMRI) showed that the functional activity of the medial prefrontal cortex (mPFC) and hippocampus was enhanced after cognitive training, and the results of magnetic resonance spectroscopy (MRS) showed that the neurochemical metabolism of N-acetyl aspartate (NAA) and glutamic acid (Glu) in mPFC, hippocampus and reuniens (Re) thalamic nucleus were escalated. Furthermore, the functional activity of mPFC and hippocampus was negatively correlated with the escape latency in memory generalization test. Therefore, these results suggested that cognitive training might improve memory generalization through enhancing the functional activity of mPFC and hippocampus and increasing the metabolism of NAA and Glu in the brain regions of mPFC, hippocampus and Re nucleus.
Collapse
Affiliation(s)
- Weilin Liu
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jianhong Li
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Le Li
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yuhao Zhang
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Minguang Yang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shengxiang Liang
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Long Li
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yaling Dai
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lewen Chen
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Weiwei Jia
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiaojun He
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Huawei Lin
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- *Correspondence: Jing Tao,
| |
Collapse
|
8
|
Neuroimaging of Mouse Models of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10020305. [PMID: 35203515 PMCID: PMC8869427 DOI: 10.3390/biomedicines10020305] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
Magnetic resonance imaging (MRI) and positron emission tomography (PET) have made great strides in the diagnosis and our understanding of Alzheimer’s Disease (AD). Despite the knowledge gained from human studies, mouse models have and continue to play an important role in deciphering the cellular and molecular evolution of AD. MRI and PET are now being increasingly used to investigate neuroimaging features in mouse models and provide the basis for rapid translation to the clinical setting. Here, we provide an overview of the human MRI and PET imaging landscape as a prelude to an in-depth review of preclinical imaging in mice. A broad range of mouse models recapitulate certain aspects of the human AD, but no single model simulates the human disease spectrum. We focused on the two of the most popular mouse models, the 3xTg-AD and the 5xFAD models, and we summarized all known published MRI and PET imaging data, including contrasting findings. The goal of this review is to provide the reader with broad framework to guide future studies in existing and future mouse models of AD. We also highlight aspects of MRI and PET imaging that could be improved to increase rigor and reproducibility in future imaging studies.
Collapse
|
9
|
Ni R. Magnetic Resonance Imaging in Animal Models of Alzheimer's Disease Amyloidosis. Int J Mol Sci 2021; 22:12768. [PMID: 34884573 PMCID: PMC8657987 DOI: 10.3390/ijms222312768] [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: 10/08/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Amyloid-beta (Aβ) plays an important role in the pathogenesis of Alzheimer's disease. Aberrant Aβ accumulation induces neuroinflammation, cerebrovascular alterations, and synaptic deficits, leading to cognitive impairment. Animal models recapitulating the Aβ pathology, such as transgenic, knock-in mouse and rat models, have facilitated the understanding of disease mechanisms and the development of therapeutics targeting Aβ. There is a rapid advance in high-field MRI in small animals. Versatile high-field magnetic resonance imaging (MRI) sequences, such as diffusion tensor imaging, arterial spin labeling, resting-state functional MRI, anatomical MRI, and MR spectroscopy, as well as contrast agents, have been developed for preclinical imaging in animal models. These tools have enabled high-resolution in vivo structural, functional, and molecular readouts with a whole-brain field of view. MRI has been used to visualize non-invasively the Aβ deposits, synaptic deficits, regional brain atrophy, impairment in white matter integrity, functional connectivity, and cerebrovascular and glymphatic system in animal models of Alzheimer's disease amyloidosis. Many of the readouts are translational toward clinical MRI applications in patients with Alzheimer's disease. In this review, we summarize the recent advances in MRI for visualizing the pathophysiology in amyloidosis animal models. We discuss the outstanding challenges in brain imaging using MRI in small animals and propose future outlook in visualizing Aβ-related alterations in the brains of animal models.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
10
|
Han H, Wang F, Chen J, Li X, Fu G, Zhou J, Zhou D, Wu W, Chen H. Changes in Biothiol Levels Are Closely Associated with Alzheimer's Disease. J Alzheimers Dis 2021; 82:527-540. [PMID: 34024827 DOI: 10.3233/jad-210021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Serum homocysteine (Hcy) level is considered to be an important biomarker for Alzheimer's disease (AD); however, the status of Hcy in brain tissue, and the association between brain and serum levels of Hcy in AD patients remain unclear. OBJECTIVE We aimed to examine whether the changes of three thiols are consistent in serum of AD patients and the brain of APP/PS1 mice, and to verify the effectiveness of Hcy as a biomarker for early AD detection. METHODS The levels of Hcy, cysteine (Cys), and glutathione (GSH) in Aβ1-42-treated PC12 cells, the brain and hippocampus of APP/PS1 mouse, and the serum of AD patients were evaluated using ethyl (E)-3-(9-chloro-11-oxo-2,3,6,7-tetrahydro-1H,5H,11H-pyrano[2,3-f] pyrido [3,2,1 -ij] quinolin-10-yl)-2-cyanoacrylate (Probe 1) and ELISA assay or LC-MS. RESULTS Measurement by Probe 1 revealed a significant increase in Hcy level, and a decrease in Cys and GSH levels in Aβ1-42-treated PC12 cells and the serum of AD patients. The hippocampus and whole brain of APP/PS1 mice also showed a significant increase in Hcy level alongside the accumulation of age-related AD symptoms. The upregulation of Hcy and the downregulation of Cys and GSH were reversed in the Aβ1-42-treated PC12 cells and the brain of APP/PS1 mice when supplemented with VB6. CONCLUSION Changes in Hcy, Cys, and GSH levels in the brain of APP/PS1 mice and Aβ1-42-treated PC12 cells were observed in situ with a new fluorescent probe, which were consistent with the abnormal changes in Hcy, Cys, and GSH levels in the serum of AD patients. VB6 supplementation was successful in ameliorating abnormal increases in Hcy levels.
Collapse
Affiliation(s)
- Hui Han
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang, China
| | - Feng Wang
- Department of Laboratory Medicine, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, China
| | - Juanjuan Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang, China
| | - Xingxing Li
- Ningbo Kangning Hospital, Ningbo Key Laboratory of Sleep Medicine, Ningbo, China
| | - Gaoqing Fu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang, China
| | - Jiawei Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang, China
| | - Dongsheng Zhou
- Ningbo Kangning Hospital, Ningbo Key Laboratory of Sleep Medicine, Ningbo, China
| | - Wei Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang, China
| | - Haimin Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
11
|
Müller L, Kirschstein T, Köhling R, Kuhla A, Teipel S. Neuronal Hyperexcitability in APPSWE/PS1dE9 Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2021; 81:855-869. [PMID: 33843674 DOI: 10.3233/jad-201540] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transgenic mouse models serve a better understanding of Alzheimer's disease (AD) pathogenesis and its consequences on neuronal function. Well-known and broadly used AD models are APPswe/PS1dE9 mice, which are able to reproduce features of amyloid-β (Aβ) plaque formations as well as neuronal dysfunction as reflected in electrophysiological recordings of neuronal hyperexcitability. The most prominent findings include abnormal synaptic function and synaptic reorganization as well as changes in membrane threshold and spontaneous neuronal firing activities leading to generalized excitation-inhibition imbalances in larger neuronal circuits and networks. Importantly, these findings in APPswe/PS1dE9 mice are at least partly consistent with results of electrophysiological studies in humans with sporadic AD. This underscores the potential to transfer mechanistic insights into amyloid related neuronal dysfunction from animal models to humans. This is of high relevance for targeted downstream interventions into neuronal hyperexcitability, for example based on repurposing of existing antiepileptic drugs, as well as the use of combinations of imaging and electrophysiological readouts to monitor effects of upstream interventions into amyloid build-up and processing on neuronal function in animal models and human studies. This article gives an overview on the pathogenic and methodological basis for recording of neuronal hyperexcitability in AD mouse models and on key findings in APPswe/PS1dE9 mice. We point at several instances to the translational perspective into clinical intervention and observation studies in humans. We particularly focus on bi-directional relations between hyperexcitability and cerebral amyloidosis, including build-up as well as clearance of amyloid, possibly related to sleep and so called glymphatic system function.
Collapse
Affiliation(s)
- Luisa Müller
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Angela Kuhla
- Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Rostock and Greifswald, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| |
Collapse
|
12
|
Zeng HM, Han HB, Zhang QF, Bai H. Application of modern neuroimaging technology in the diagnosis and study of Alzheimer's disease. Neural Regen Res 2021; 16:73-79. [PMID: 32788450 PMCID: PMC7818875 DOI: 10.4103/1673-5374.286957] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/14/2020] [Accepted: 03/28/2020] [Indexed: 12/13/2022] Open
Abstract
Neurological abnormalities identified via neuroimaging are common in patients with Alzheimer's disease. However, it is not yet possible to easily detect these abnormalities using head computed tomography in the early stages of the disease. In this review, we evaluated the ways in which modern imaging techniques such as positron emission computed tomography, single photon emission tomography, magnetic resonance spectrum imaging, structural magnetic resonance imaging, magnetic resonance diffusion tensor imaging, magnetic resonance perfusion weighted imaging, magnetic resonance sensitive weighted imaging, and functional magnetic resonance imaging have revealed specific changes not only in brain structure, but also in brain function in Alzheimer's disease patients. The reviewed literature indicated that decreased fluorodeoxyglucose metabolism in the temporal and parietal lobes of Alzheimer's disease patients is frequently observed via positron emission computed tomography. Furthermore, patients with Alzheimer's disease often show a decreased N-acetylaspartic acid/creatine ratio and an increased myoinositol/creatine ratio revealed via magnetic resonance imaging. Atrophy of the entorhinal cortex, hippocampus, and posterior cingulate gyrus can be detected early using structural magnetic resonance imaging. Magnetic resonance sensitive weighted imaging can show small bleeds and abnormal iron metabolism. Task-related functional magnetic resonance imaging can display brain function activity through cerebral blood oxygenation. Resting functional magnetic resonance imaging can display the functional connection between brain neural networks. These are helpful for the differential diagnosis and experimental study of Alzheimer's disease, and are valuable for exploring the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Hong-Mei Zeng
- Department of Neurology, Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou Province, China
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hua-Bo Han
- Department of Radiology, Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou Province, China
| | - Qi-Fang Zhang
- Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, and Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hua Bai
- Department of Neurology, Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou Province, China
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
- Medical Experiment Center, Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou Province, China
| |
Collapse
|
13
|
Müller HP, Roselli F, Rasche V, Kassubek J. Diffusion Tensor Imaging-Based Studies at the Group-Level Applied to Animal Models of Neurodegenerative Diseases. Front Neurosci 2020; 14:734. [PMID: 32982659 PMCID: PMC7487414 DOI: 10.3389/fnins.2020.00734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
The understanding of human and non-human microstructural brain alterations in the course of neurodegenerative diseases has substantially improved by the non-invasive magnetic resonance imaging (MRI) technique of diffusion tensor imaging (DTI). Animal models (including disease or knockout models) allow for a variety of experimental manipulations, which are not applicable to humans. Thus, the DTI approach provides a promising tool for cross-species cross-sectional and longitudinal investigations of the neurobiological targets and mechanisms of neurodegeneration. This overview with a systematic review focuses on the principles of DTI analysis as used in studies at the group level in living preclinical models of neurodegeneration. The translational aspect from in-vivo animal models toward (clinical) applications in humans is covered as well as the DTI-based research of the non-human brains' microstructure, the methodological aspects in data processing and analysis, and data interpretation at different abstraction levels. The aim of integrating DTI in multiparametric or multimodal imaging protocols will allow the interrogation of DTI data in terms of directional flow of information and may identify the microstructural underpinnings of neurodegeneration-related patterns.
Collapse
Affiliation(s)
| | - Francesco Roselli
- Department of Neurology, University of Ulm, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Volker Rasche
- Core Facility Small Animal MRI, University of Ulm, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, University of Ulm, Ulm, Germany
| |
Collapse
|
14
|
Wang L, Zeng L, Jiang H, Li Z, Liu R. Microarray Profile of Long Noncoding RNA and Messenger RNA Expression in a Model of Alzheimer's Disease. Life (Basel) 2020; 10:E64. [PMID: 32423012 PMCID: PMC7281340 DOI: 10.3390/life10050064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by a deficiency in cognitive skills. Although long noncoding RNAs (lncRNAs) have been proposed as associated with AD, the aberrant lncRNAs expression and the co-expression of lncRNAs-mRNAs network in AD remains unclear. Therefore, in this study, lncRNA microarray was performed on the brain of APP/PS1 mice at different age, widely used as an AD mouse model, and on age-matched wide-type controls. Our results identified a total of 3306 lncRNAs and 2458 mRNAs as aberrantly expressed among AD mice at different age and their age-matched control. Gene Ontology and pathway analysis of the AD-related lncRNAs and mRNAs indicated that neuroinflammation-related and synaptic transmission signaling pathways represented the main enriched pathways. An lncRNA-mRNA-miRNA network between the differentially expressed transcripts was constructed. Moreover, an mRNA-miRNA network between both significantly dysregulated and highly conserved genes was also constructed, and among this network, the IGF1, P2RX7, TSPO, SERPINE1, EGFR, HMOX1, and NFE212 genes were predicted to play a role in the development of AD. In conclusion, this study illustrated the prognostic value of lncRNAs and mRNAs associated to AD pathology by microarray analysis and might provide potential novel biomarkers in the diagnosis and treatment of AD.
Collapse
Affiliation(s)
| | | | | | - Zhuorong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (L.W.); (L.Z.); (H.J.)
| | - Rui Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (L.W.); (L.Z.); (H.J.)
| |
Collapse
|
15
|
Pilot study utilizing MRI 3D TGSE PASL (arterial spin labeling) differentiating clearance rates of labeled protons in the CNS of patients with early Alzheimer disease from normal subjects. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2020; 33:559-568. [DOI: 10.1007/s10334-019-00818-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 01/21/2023]
|
16
|
Genomic deletion of TLR2 induces aggravated white matter damage and deteriorated neurobehavioral functions in mouse models of Alzheimer's disease. Aging (Albany NY) 2019; 11:7257-7273. [PMID: 31509519 PMCID: PMC6756907 DOI: 10.18632/aging.102260] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 09/02/2019] [Indexed: 01/09/2023]
Abstract
Toll-like receptor-2 (TLR2), a member of the TLR family, plays an important role in the initiation and regulation of immune/inflammation response, which is a critical mechanism underlying Alzheimer’s disease (AD). To clarify the role of TLR2 in the pathological process of AD, in the present study, TLR2 knockout plus APPswe/PSEN1dE9 transgenic mice (AD-TLR2KO) were generated. Neurobehavioral tests and brain MRI scan were conducted on mice at the age of 12 months. Additionally, neuron loss was evaluated using NeuN staining. Amyloid β protein (Aβ), glial fibrillary acidic protein (GFAP), endogenous ligands for TLR2, and the activation of downstream signaling of TLR2 in mouse brains were detected by immunohistochemistry and Western blots. The results demonstrated that TLR2 deficit induced learning disabilities, decreased spontaneous activity, increased anxiety and depression, and led to white matter damage (WMD), brain atrophy, loss of neurons, and glial activation. Moreover, TLR2 deficit aggravated impaired neurobehavioral functions and WMD in AD mice, but did not affect the Aβ deposition in mouse brains. Our data indicate that the genomic deletion of TLR2 impairs neurobehavioral functions, induces WMD and brain atrophy, and increases the activation of astrocytes, which in turn aggravate the symptoms of AD through a non-Aβ mechanism.
Collapse
|
17
|
Zhang X, Yin X, Zhang J, Li A, Gong H, Luo Q, Zhang H, Gao Z, Jiang H. High-resolution mapping of brain vasculature and its impairment in the hippocampus of Alzheimer's disease mice. Natl Sci Rev 2019; 6:1223-1238. [PMID: 34692000 PMCID: PMC8291402 DOI: 10.1093/nsr/nwz124] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 01/24/2023] Open
Abstract
Accumulating evidence indicates the critical importance of cerebrovascular dysfunction in the pathogenesis of Alzheimer's disease (AD). However, systematic comparative studies on the precise brain vasculature of wild-type and AD model mice are still rare. Using an image-optimization method for analysing Micro-Optical Sectioning Tomography (MOST) data, we generated cross-scale whole-brain 3D atlases that cover the entire vascular system from large vessels down to smallest capillaries at submicron resolution, for both wild-type mice and a transgenic (APP/PS1) mouse model of AD. In addition to distinct vascular patterns in different brain regions, we found that the main vessels of the molecular layer of the hippocampal dentate gyrus (DG-ml) undergo abrupt changes in both diameter and branch angle, spreading a unique comb-like pattern of capillaries. By using a quantitative analysis workflow, we identified in the hippocampus of AD mice an overall reduction of the mean vascular diameter, volume fraction and branch angle, with most significant impairment in the DG-ml. In addition, virtual endoscopy revealed irregular morphological features in the vessel lumen of the AD mice, potentially contributing to the impairment of blood flow. Our results demonstrate the capability of high-resolution cross-scale evaluation of brain vasculature and underscore the importance of studying hippocampal microcirculation for understanding AD pathogenesis.
Collapse
Affiliation(s)
- Xiaochuan Zhang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xianzhen Yin
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingjing Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Haiyan Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhaobing Gao
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hualiang Jiang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute for Advanced Immunochemical Studies, and School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| |
Collapse
|
18
|
Sun H, Liu M, Sun T, Chen Y, Lan Z, Lian B, Zhao C, Liu Z, Zhang J, Liu Y. Age-related changes in hippocampal AD pathology, actin remodeling proteins and spatial memory behavior of male APP/PS1 mice. Behav Brain Res 2019; 376:112182. [PMID: 31472195 DOI: 10.1016/j.bbr.2019.112182] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly, characterized by amyloid-beta (Aβ) plaques and tau neurofibrillary tangles (NFTs). Synaptic plasticity impairment is one of the early pathological events in AD. Transgenic APP/PS1 mice that overproduce Aβ are one of the most extensively used AD animal models. Many studies have investigated the roles of NTF-related p-Tau, non-amyloidogenic ADAM10, amyloidogenic BACE1, Aβ proteolytic NEP and IDE in certain ages of APP/PS1 mice as well as dendritic spine-related Rictor and Profilin-1 in normal mice, but there are few studies exploring the age-related changes of these molecules in the hippocampus of APP/PS1 mice. Furthermore, current studies regarding when memory impairment occurs in these mice are controversial. Thus, we examined the changes of these molecules in APP/PS1 and control mice using Western blot in mice 2-month-old (2 m) to 10 m of age and behavior changes using the Morris water maze from 4 m to 8 m. The results showed that in APP/PS1 mice, significant changes of hippocampal p-Tau, Aβ, ADAM10, BACE1 and Rictor occurred at 6 m, NEP at 8 m, and IDE and Profilin-1 at 10 m. In control mice, changes of p-Tau, ADAM10, and BACE1 occurred at 8 m and NEP at 10 m, while IDE, Rictor and Profilin-1 remained unchanged. Importantly, the Morris water maze test revealed that spatial memory impairment was detected at 8 m but not 4 or 6 m. The above findings clearly evidence that neurochemical changes overtly precede cognitive dysfunctions in this AD model and provide novel knowledge for a better understanding of the molecular events driving AD.
Collapse
Affiliation(s)
- Huan Sun
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Mengying Liu
- Department of Neurobiology, Third Military Medical University, Chongqing, 400038, China
| | - Tao Sun
- Department of Neurobiology, Third Military Medical University, Chongqing, 400038, China
| | - Yutong Chen
- Student Brigade, College of Basic Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Zhen Lan
- Department of Neurobiology, Third Military Medical University, Chongqing, 400038, China
| | - Biyao Lian
- Department of Histology and Embryology, Ningxia Medical University, Yinchuan, 750004, China
| | - Chengjun Zhao
- Department of Histology and Embryology, Ningxia Medical University, Yinchuan, 750004, China
| | - Zhi Liu
- Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Jiqiang Zhang
- Department of Neurobiology, Third Military Medical University, Chongqing, 400038, China.
| | - Yan Liu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|