1
|
Darwish AB, Salama A, Younis MM. Neuroprotective efficiency of celecoxib vesicular bilosomes for the management of lipopolysaccharide-induced Alzheimer in mice employing 2 3 full factorial design. Inflammopharmacology 2024; 32:3925-3942. [PMID: 39017993 PMCID: PMC11550292 DOI: 10.1007/s10787-024-01522-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/02/2024] [Indexed: 07/18/2024]
Abstract
The aim of this study was to develop and evaluate bilosomes loaded with Celecoxib (CXB) for the efficient treatment of Alzheimer. The thin-film hydration approach was utilized in the formulation of CXB bilosomes (CXB-BLs). The study used a 23-factorial design to investigate the impact of several formulation variables. Three separate parameters were investigated: bile salt type (X1), medication amount (X2), and lipid-bile salt ratio (X3). The dependent responses included entrapment efficiency (Y1: EE %), particle size (Y2: PS), and zeta potential (Y3: ZP). The formulation factors were statistically optimized using the Design-Expert® program. The vesicles demonstrated remarkable CXB encapsulation efficiency, ranging from 94.16 ± 1.91 to 98.38 ± 0.85%. The vesicle sizes ranged from 241.8 ± 6.74 to 352 ± 2.34 nm. The produced formulations have high negative zeta potential values, indicating strong stability. Transmission electron microscopy (TEM) revealed that the optimized vesicles had a spherical form. CXB release from BLs was biphasic, with the release pattern following Higuchi's model. In vivo studies confirmed the efficiency of CXB-BLs in management of lipopolysaccharide-induced Alzheimer as CXB-BLs ameliorated cognitive dysfunction, decreased acetylcholinesterase (AChE), and inhibited neuro-inflammation and neuro-degeneration through reducing Toll-like receptor (TLR4), and Interleukin-1β (IL-1β) levels. The findings suggested that the created CXB-BLs could be a potential drug delivery strategy for Alzheimer's treatment.
Collapse
Affiliation(s)
- Asmaa Badawy Darwish
- Pharmaceutical Technology Department, National Research Centre, 33 El-Buhouth Street, Dokki, Cairo, 12622, Egypt.
| | - Abeer Salama
- Pharmacology Department, National Research Centre, 33 El-Buhouth Street, Dokki, Cairo, 12622, Egypt
| | - Mostafa Mohammed Younis
- Pharmaceutical Technology Department, National Research Centre, 33 El-Buhouth Street, Dokki, Cairo, 12622, Egypt
| |
Collapse
|
2
|
Zhou X, Liu Y, Wu Z, Zhang X, Tao H. Alzheimer's disease and epilepsy: Research hotspots for comorbidity in the era of global aging. Epilepsy Behav 2024; 157:109849. [PMID: 38820684 DOI: 10.1016/j.yebeh.2024.109849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024]
Abstract
Neurological conditions such as Alzheimer's disease (AD) and epilepsy share a significant clinical overlap, particularly in the elderly, with each disorder potentiating the risk of the other. This interplay is significant amidst an aging global demographic. The review explores the classical pathologies of AD, including amyloid-beta plaques and hyperphosphorylated tau, and their potential role in the genesis of epilepsy. It also delves into the imbalance of glutamate and gamma-amino butyric acid activities, a key mechanism in epilepsy that may be influenced by AD pathology. The impact of age of onset on comorbidity is examined, with early-onset AD and Down syndrome presenting higher risks of epilepsy. The review suggests that epilepsy might precede cognitive symptoms in AD, indicating a complex interaction. Sleep modulation is highlighted as a factor, with sleep disturbances potentially contributing to AD progression. The necessity for cautious medication management is emphasized due to the cognitive effects of certain antiepileptic drugs. Animal models are recognized for their importance in understanding the relationship between AD and epilepsy, though creating fully representative models presents a challenge. The review concludes by noting the efficacy of medications such as lamotrigine, levetiracetam, and memantine in managing both conditions and suggests the ketogenic diet and cannabidiol as emerging treatment options, warranting further investigation for comprehensive patient care strategies.
Collapse
Affiliation(s)
- Xu Zhou
- Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Yang Liu
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Zhengjuan Wu
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Xiaolu Zhang
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Hua Tao
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Guangdong Key Laboratory of Age-related Cardiac and Cerebral Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, China.
| |
Collapse
|
3
|
Song C, Li S, Mai Y, Li L, Dai G, Zhou Y, Liang X, Zou OM, Wang Y, Zhou L, Liu J, Zou Y. Dysregulated expression of miR-140 and miR-122 compromised microglial chemotaxis and led to reduced restriction of AD pathology. J Neuroinflammation 2024; 21:167. [PMID: 38956605 PMCID: PMC11218311 DOI: 10.1186/s12974-024-03162-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Deposition of amyloid β, which is produced by amyloidogenic cleavage of APP by β- and γ-secretase, is one of the primary hallmarks of AD pathology. APP can also be processed by α- and γ-secretase sequentially, to generate sAPPα, which has been shown to be neuroprotective by promoting neurite outgrowth and neuronal survival, etc. METHODS: The global expression profiles of miRNA in blood plasma samples taken from 11 AD patients as well as from 14 age and sex matched cognitively normal volunteers were analyzed using miRNA-seq. Then, overexpressed miR-140 and miR-122 both in vivo and in vitro, and knock-down of the endogenous expression of miR-140 and miR-122 in vitro. Used a combination of techniques, including molecular biology, immunohistochemistry, to detect the impact of miRNAs on AD pathology. RESULTS In this study, we identified that two miRNAs, miR-140-3p and miR-122-5p, both targeting ADAM10, the main α-secretase in CNS, were upregulated in the blood plasma of AD patients. Overexpression of these two miRNAs in mouse brains induced cognitive decline in wild type C57BL/6J mice as well as exacerbated dyscognition in APP/PS1 mice. Although significant changes in APP and total Aβ were not detected, significantly downregulated ADAM10 and its non-amyloidogenic product, sAPPα, were observed in the mouse brains overexpressing miR-140/miR-122. Immunohistology analysis revealed increased neurite dystrophy that correlated with the reduced microglial chemotaxis in the hippocampi of these mice, independent of the other two ADAM10 substrates (neuronal CX3CL1 and microglial TREM2) that were involved in regulating the microglial immunoactivity. Further in vitro analysis demonstrated that both the reduced neuritic outgrowth of mouse embryonic neuronal cells overexpressing miR-140/miR-122 and the reduced Aβ phagocytosis in microglia cells co-cultured with HT22 cells overexpressing miR-140/miR-122 could be rescued by overexpressing the specific inhibitory sequence of miR-140/miR-122 TuD as well as by addition of sAPPα, rendering these miRNAs as potential therapeutic targets. CONCLUSIONS Our results suggested that neuroprotective sAPPα was a key player in the neuropathological progression induced by dysregulated expression of miR-140 and miR-122. Targeting these miRNAs might serve as a promising therapeutic strategy in AD treatment.
Collapse
Affiliation(s)
- Chao Song
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Shufang Li
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Yingren Mai
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Linpeng Li
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Guoku Dai
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Yuan Zhou
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xiaosheng Liang
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Olivia Meilan Zou
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Ya Wang
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Jun Liu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
| | - Yi Zou
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China.
| |
Collapse
|
4
|
Gao L, Wang D, Ren J, Tan X, Chen J, Kong Z, Nie Y, Yan M. Acteoside ameliorates learning and memory impairment in APP/PS1 transgenic mice by increasing Aβ degradation and inhibiting tau hyperphosphorylation. Phytother Res 2024; 38:1735-1744. [PMID: 37661763 DOI: 10.1002/ptr.8006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 06/27/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease. Senile plaques and intracellular neurofibrillary tangles are pathological hallmarks of AD. Recent studies have described the improved cognitive and neuroprotective functions of acteoside (AS). This study aimed to investigate whether the improved cognition of AS was mediated by Aβ degradation and tau phosphorylation in APP/PS1 mice. The open field, Y maze, and novel object recognition tests were used to assess cognitive behavioral changes. We evaluated the levels of Aβ40 and Aβ42 in serum, cortex, and hippocampus, and Aβ-related scavenging enzymes, phosphorylated GSK3β and hyperphosphorylated tau in the cortex and hippocampus of APP/PS1 mice by western blotting. Our results revealed that AS treatment ameliorated anxious behaviors, spatial learning, and memory impairment in APP/PS1 mice and significantly reduced Aβ deposition in their serum, cortex, and hippocampus. AS significantly increased Aβ degradation, inhibited the hyperphosphorylation of tau, and significantly decreased the activity of GSK3β, which is involved in tau phosphorylation. Altogether, these findings indicated that the beneficial effects of AS on AD-associated anxious behaviors and cognitive impairments could be attributed to promoting Aβ degradation and inhibiting tau hyperphosphorylation, which might be partly mediated by GSK3β.
Collapse
Affiliation(s)
- Li Gao
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Dongqing Wang
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Jia Ren
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Xue Tan
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Jiayuan Chen
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Zheng Kong
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Yunan Nie
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| | - Ming Yan
- Prescription Laboratory of Xinjiang Traditional Uyghur Medicine, Xinjiang Institute of Traditional Uyghur Medicine, Urumqi, China
| |
Collapse
|
5
|
Fessel J. Personalized, Precision Medicine to Cure Alzheimer's Dementia: Approach #1. Int J Mol Sci 2024; 25:3909. [PMID: 38612719 PMCID: PMC11012190 DOI: 10.3390/ijms25073909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
The goal of the treatment for Alzheimer's dementia (AD) is the cure of dementia. A literature review revealed 18 major elements causing AD and 29 separate medications that address them. For any individual with AD, one is unlikely to discern which major causal elements produced dementia. Thus, for personalized, precision medicine, all causal elements must be treated so that each individual patient will have her or his causal elements addressed. Twenty-nine drugs cannot concomitantly be administered, so triple combinations of drugs taken from that list are suggested, and each triple combination can be administered sequentially, in any order. Ten combinations given over 13 weeks require 2.5 years, or if given over 26 weeks, they require 5.0 years. Such sequential treatment addresses all 18 elements and should cure dementia. In addition, any comorbid risk factors for AD whose first presence or worsening was within ±1 year of when AD first appeared should receive appropriate, standard treatment together with the sequential combinations. The article outlines a randomized clinical trial that is necessary to assess the safety and efficacy of the proposed treatments; it includes a triple-drug Rx for equipoise. Clinical trials should have durations of both 2.5 and 5.0 years unless the data safety monitoring board (DSMB) determines earlier success or futility since it is uncertain whether three or six months of treatment will be curative in humans, although studies in animals suggest that the briefer duration of treatment might be effective and restore defective neural tracts.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Clinical Medicine, Department of Medicine, University of California, 2069 Filbert Street, San Francisco, CA 94123, USA
| |
Collapse
|
6
|
Feizolahi F, Arabzadeh E, Sarshin A, Falahi F, Dehghannayeri Z, Ali Askari A, Wong A, Aghaei F, Zargani M. Effects of Exercise Training and L-Arginine Loaded Chitosan Nanoparticles on Hippocampus Histopathology, β-Secretase Enzyme Function, APP, Tau, Iba1and APOE-4 mRNA in Aging Rats. Neurotox Res 2024; 42:21. [PMID: 38441819 DOI: 10.1007/s12640-024-00699-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024]
Abstract
The objective of this study was to evaluate the combined and independent effects of exercise training and L-Arginine loaded chitosan nanoparticles (LA CNPs) supplementation on hippocampal Tau, App, Iba1, and ApoE gene expression, oxidative stress, β-secretase enzyme activity, and hippocampus histopathology in aging rats. Thirty-five male Wistar rats were randomly assigned to five groups (n = 7 in each): Young (8 weeks old), Old (20 months old), old + L-arginine supplementation (Old Sup), old + exercise (Old Exe) and old + L-arginine supplementation + exercise (Old Sup + Exe). LA CNPs were administered to the supplement groups through gavage at a dosage of 500 mg/kg/day for 6-weeks. Exercise groups were subjected to a swimming exercise program five days/week for the same duration. Upon the completion of their interventions, the animals underwent behavioral and open-field task tests and were subsequently sacrificed for hippocampus genetic and histopathological evaluation. For histopathological analysis of brain, Cresyl violet staining was used. Congo Red staining was employed to confirm amyloid plaques in the hippocampus. Expressions of Tau, App, Iba1, and ApoE genes were determined by real-time PCR. In contrast to the Old group, Old Exe and Old Sup + Exe groups spent more time in the central space in the open field task (p < 0.05) and have more live cells in the hippocampus. Old rats (Old, Old Sup and Old Exe groups) exhibited a significant Aβ peptide accumulation and increases in APP, Tau, Iba1, APOE-4 mRNA and MDA, along with decreases in SOD compared to the young group (p < 0.05). However, LA CNPs supplementation, exercise, and their combination (Old Sup, Old Exe and Old Sup + Exe) significantly reduced MDA, Aβ plaque as well as APP, Tau, Iba1, and APOE-4 mRNA compared to the Old group (p < 0.05). Consequently, the administration of LA CNPs supplements and exercise might regulate the risk factors of hippocampus cell and tissue.
Collapse
Affiliation(s)
- Foad Feizolahi
- Clinical Care and Health Promotion Research Center, Karaj branch, Islamic Azad University, Karaj, Iran
| | - Ehsan Arabzadeh
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir Sarshin
- Clinical Care and Health Promotion Research Center, Karaj branch, Islamic Azad University, Karaj, Iran
| | - Farshad Falahi
- Department of Exercise Physiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Zahra Dehghannayeri
- Department of Exercise Physiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Ali Ali Askari
- Department of Exercise Physiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Alexei Wong
- Department of Health and Human Performance, Marymount University, Arlington, VA, USA
| | - Fariba Aghaei
- Department of Exercise Physiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Mehdi Zargani
- Department of Exercise Physiology, Karaj Branch, Islamic Azad University, Karaj, Iran.
| |
Collapse
|
7
|
He J, Liu Y, Li J, Zhao Y, Jiang H, Luo S, He G. Intestinal changes in permeability, tight junction and mucin synthesis in a mouse model of Alzheimer's disease. Int J Mol Med 2023; 52:113. [PMID: 37830152 PMCID: PMC10599350 DOI: 10.3892/ijmm.2023.5316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/11/2023] [Indexed: 10/14/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid‑β (Aβ) in the brain. The gut/brain axis may serve a role in AD pathogenesis. The present study investigated deposition of Aβ in the intestinal epithelium and its potential effects on intestinal barrier function in a transgenic mouse model of AD. To investigate alterations in the structure and functionality of the intestinal mucosal barrier in AD model mice, hematoxylin and eosin staining for Paneth cell count, Alcian blue‑periodic acid Schiff staining for goblet cells, immunohistochemistry and immunofluorescence for mucin (MUC)2 and wheat germ agglutin expression, transmission electron microscopy for mucosal ultrastructure, FITC‑labeled dextran assay for intestinal permeability, quantitative PCR for goblet cell precursor expression and western blot analysis for tight junction proteins, MUC2 and inflammatory cytokine detection were performed. The results showed that AD model mice exhibited excessive Aβ deposition in the intestinal epithelium, which was accompanied by increased intestinal permeability, inflammatory changes and decreased expression of tight junction proteins. These alterations in the intestinal barrier led to an increased proliferation of goblet and Paneth cells and increased mucus synthesis. Dysfunction of gut barrier occurs in AD and may contribute to its etiology. Future therapeutic strategies to reverse AD pathology may involve early manipulation of gut physiology and its microbiota.
Collapse
Affiliation(s)
- Jing He
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
- Department of Anatomy, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
| | - Yuanjie Liu
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
- Department of Anatomy, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
| | - Junhua Li
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
- Department of Anatomy, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
| | - Yueyang Zhao
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
- Department of Anatomy, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
| | - Hanxiao Jiang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Shifang Luo
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
- Department of Anatomy, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
| | - Guiqiong He
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
- Department of Anatomy, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016
| |
Collapse
|
8
|
Kepp KP, Robakis NK, Høilund-Carlsen PF, Sensi SL, Vissel B. The amyloid cascade hypothesis: an updated critical review. Brain 2023; 146:3969-3990. [PMID: 37183523 DOI: 10.1093/brain/awad159] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/16/2023] Open
Abstract
Results from recent clinical trials of antibodies that target amyloid-β (Aβ) for Alzheimer's disease have created excitement and have been heralded as corroboration of the amyloid cascade hypothesis. However, while Aβ may contribute to disease, genetic, clinical, imaging and biochemical data suggest a more complex aetiology. Here we review the history and weaknesses of the amyloid cascade hypothesis in view of the new evidence obtained from clinical trials of anti-amyloid antibodies. These trials indicate that the treatments have either no or uncertain clinical effect on cognition. Despite the importance of amyloid in the definition of Alzheimer's disease, we argue that the data point to Aβ playing a minor aetiological role. We also discuss data suggesting that the concerted activity of many pathogenic factors contribute to Alzheimer's disease and propose that evolving multi-factor disease models will better underpin the search for more effective strategies to treat the disease.
Collapse
Affiliation(s)
- Kasper P Kepp
- Section of Biophysical and Biomedicinal chemistry, DTU Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Nikolaos K Robakis
- Icahn School of Medicine at Mount Sinai Medical Center, New York, NY 10029, USA
| | - Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Stefano L Sensi
- Center for Advanced Studies and Technology-CAST, and Institute for Advanced Biotechnology (ITAB), University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
| | - Bryce Vissel
- St Vincent's Hospital Centre for Applied Medical Research, St Vincent's Hospital, Sydney, 2010, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, Sydney, NSW 2052, Australia
| |
Collapse
|
9
|
Liu GZ, Niu TT, Yu Q, Xu BL, Li XQ, Yuan BY, Yuan GB, Yang TT, Li HQ, Sun Y. Ginkgolide attenuates memory impairment and neuroinflammation by suppressing the NLRP3/caspase-1 pathway in Alzheimer's disease. Aging (Albany NY) 2023; 15:10237-10252. [PMID: 37793010 PMCID: PMC10599747 DOI: 10.18632/aging.205072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/17/2023] [Indexed: 10/06/2023]
Abstract
The NLRP3 inflammasome is involved in the neuroinflammatory pathway of Alzheimer's disease (AD). The aim of this study is to explore the roles and underlying mechanisms of ginkgolide (Baiyu®) on amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mice and a murine microglial cell line, BV-2. In the present study, the APP/PS1 mice were administered with ginkgolide, followed by a Morris water maze test. The mice were then euthanized to obtain brain tissue for histological and Aβ analysis. Additionally, BV-2 cells were pretreated with ginkgolide and then incubated with Aβ1-42 peptide. NLRP3, ASC, and caspase-1 mRNA and protein expression in brain tissue of mice and BV-2 cells were quantified by real-time PCR and western blotting, as well as reactive oxygen species (ROS) production, interleukin (IL)-1β and IL-18 levels by lucigenin technique and ELISA. Compared with the APP/PS1 mice, ginkgolide-treated mice demonstrated the shortened escape latency, reduced plaques, less inflammatory cell infiltration and neuron loss in the hippocampi of APP/PS1 mice. The levels of NLRP3, ASC, caspase-1, ROS, IL-1β, and IL-18 were also decreased in the brain tissue of APP/PS1 mice or Aβ1-42-treated BV-2 cells following ginkgolide treatment. Ginkgolide exerted protective effects on AD, at least partly by inactivating the NLRP3/caspase-1 pathway.
Collapse
Affiliation(s)
- Guang-Zhi Liu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Tian-Tong Niu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Qian Yu
- Beijing D.A. Medical Laboratory, Beijing 102600, China
| | - Bao-Lei Xu
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiao-Qing Li
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Bo-Yi Yuan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Guo-Bin Yuan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ting-Ting Yang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Hui-Qin Li
- Research and Development Centre, Chengdu Baiyu Pharmaceutical Co., Ltd., Chengdu 611130, China
| | - Yi Sun
- Research and Development Centre, Chengdu Baiyu Pharmaceutical Co., Ltd., Chengdu 611130, China
| |
Collapse
|
10
|
Bedoya-Guzmán FA, Pacheco-Herrero M, Salomon-Cruz ID, Barrera-Sandoval AM, Gutierrez Vargas JA, Villamil-Ortiz JG, Villegas Lanau CA, Arias-Londoño JD, Area-Gomez E, Cardona Gomez GP. BACE1 and SCD1 are associated with neurodegeneration. Front Aging Neurosci 2023; 15:1194203. [PMID: 37744400 PMCID: PMC10516302 DOI: 10.3389/fnagi.2023.1194203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/03/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Proteolytic processing of amyloid protein precursor by β-site secretase enzyme (BACE1) is dependent on the cellular lipid composition and is affected by endomembrane trafficking in dementia and Alzheimer's disease (AD). Stearoyl-CoA desaturase 1 (SCD1) is responsible for the synthesis of fatty acid monounsaturation (MUFAs), whose accumulation is strongly associated with cognitive dysfunction. Methods In this study, we analyzed the relationship between BACE1 and SCD1 in vivo and in vitro neurodegenerative models and their association in familial AD (FAD), sporadic AD (SAD), and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) using microscopy, biochemical, and mass SPECT approach. Results Our findings showed that BACE1 and SCD1 immunoreactivities were increased and colocalized in astrocytes of the hippocampus in a rat model of global cerebral ischemia (2-VO). A synergistic effect of double BACE1/SCD1 silencing on the recovery of motor and cognitive functions was obtained. This neuroprotective regulation involved the segregation of phospholipids (PLs) associated with polyunsaturated fatty acids in the hippocampus, cerebrospinal fluid, and serum. The double silencing in the sham and ischemic groups was stronger in the serum, inducing an inverse ratio between total phosphatydilcholine (PC) and lysophosphatidylcholine (LPC), represented mainly by the reduction of PC 38:4 and PC 36:4 and an increase in LPC 16:0 and LPC 18:0. Furthermore, PC 38:4 and PC:36:4 levels augmented in pathological conditions in in vitro AD models. BACE1 and SCD1 increases were confirmed in the hippocampus of FAD, SAD, and CADASIL. Conclusion Therefore, the findings suggest a novel convergence of BACE-1 and SCD1 in neurodegeneration, related to pro-inflammatory phospholipids.
Collapse
Affiliation(s)
- Ferley A. Bedoya-Guzmán
- Faculty of Medicine University of Antioquia, Cellular and Molecular Neurobiology Area and Neurobank, Group of Neuroscience (GNA), Medellín, Colombia
| | - Mar Pacheco-Herrero
- Faculty of Medicine University of Antioquia, Cellular and Molecular Neurobiology Area and Neurobank, Group of Neuroscience (GNA), Medellín, Colombia
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Santiago de los Caballeros, Dominican Republic
| | - Ivan Daniel Salomon-Cruz
- Faculty of Medicine University of Antioquia, Cellular and Molecular Neurobiology Area and Neurobank, Group of Neuroscience (GNA), Medellín, Colombia
| | - Angela Maria Barrera-Sandoval
- Faculty of Medicine University of Antioquia, Cellular and Molecular Neurobiology Area and Neurobank, Group of Neuroscience (GNA), Medellín, Colombia
| | - Johanna Andrea Gutierrez Vargas
- Faculty of Medicine University of Antioquia, Cellular and Molecular Neurobiology Area and Neurobank, Group of Neuroscience (GNA), Medellín, Colombia
- Grupo de Investigación en Salud del Adulto Mayor (GISAM), Corporación Universitaria Remington, Medellín, Colombia
| | - Javier Gustavo Villamil-Ortiz
- Faculty of Medicine University of Antioquia, Cellular and Molecular Neurobiology Area and Neurobank, Group of Neuroscience (GNA), Medellín, Colombia
| | - Carlos Andres Villegas Lanau
- Faculty of Medicine University of Antioquia, Cellular and Molecular Neurobiology Area and Neurobank, Group of Neuroscience (GNA), Medellín, Colombia
| | | | - Estela Area-Gomez
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| | - Gloria Patricia Cardona Gomez
- Faculty of Medicine University of Antioquia, Cellular and Molecular Neurobiology Area and Neurobank, Group of Neuroscience (GNA), Medellín, Colombia
| |
Collapse
|
11
|
Cools R, Kerkhofs K, Leitao RCF, Bormans G. Preclinical Evaluation of Novel PET Probes for Dementia. Semin Nucl Med 2023; 53:599-629. [PMID: 37149435 DOI: 10.1053/j.semnuclmed.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 05/08/2023]
Abstract
The development of novel PET imaging agents that selectively bind specific dementia-related targets can contribute significantly to accurate, differential and early diagnosis of dementia causing diseases and support the development of therapeutic agents. Consequently, in recent years there has been a growing body of literature describing the development and evaluation of potential new promising PET tracers for dementia. This review article provides a comprehensive overview of novel dementia PET probes under development, classified by their target, and pinpoints their preclinical evaluation pathway, typically involving in silico, in vitro and ex/in vivo evaluation. Specific target-associated challenges and pitfalls, requiring extensive and well-designed preclinical experimental evaluation assays to enable successful clinical translation and avoid shortcomings observed for previously developed 'well-established' dementia PET tracers are highlighted in this review.
Collapse
Affiliation(s)
- Romy Cools
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Kobe Kerkhofs
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; NURA, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Renan C F Leitao
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
12
|
Chen C, Wei J, Ma X, Xia B, Shakir N, Zhang JK, Zhang L, Cui Y, Ferguson D, Qiu S, Bai F. Disrupted Maturation of Prefrontal Layer 5 Neuronal Circuits in an Alzheimer's Mouse Model of Amyloid Deposition. Neurosci Bull 2023; 39:881-892. [PMID: 36152121 PMCID: PMC10264337 DOI: 10.1007/s12264-022-00951-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/18/2022] [Indexed: 11/26/2022] Open
Abstract
Mutations in genes encoding amyloid precursor protein (APP) and presenilins (PSs) cause familial forms of Alzheimer's disease (AD), a neurodegenerative disorder strongly associated with aging. It is currently unknown whether and how AD risks affect early brain development, and to what extent subtle synaptic pathology may occur prior to overt hallmark AD pathology. Transgenic mutant APP/PS1 over-expression mouse lines are key tools for studying the molecular mechanisms of AD pathogenesis. Among these lines, the 5XFAD mice rapidly develop key features of AD pathology and have proven utility in studying amyloid plaque formation and amyloid β (Aβ)-induced neurodegeneration. We reasoned that transgenic mutant APP/PS1 over-expression in 5XFAD mice may lead to neurodevelopmental defects in early cortical neurons, and performed detailed synaptic physiological characterization of layer 5 (L5) neurons from the prefrontal cortex (PFC) of 5XFAD and wild-type littermate controls. L5 PFC neurons from 5XFAD mice show early APP/Aβ immunolabeling. Whole-cell patch-clamp recording at an early post-weaning age (P22-30) revealed functional impairments; although 5XFAD PFC-L5 neurons exhibited similar membrane properties, they were intrinsically less excitable. In addition, these neurons received smaller amplitude and frequency of miniature excitatory synaptic inputs. These functional disturbances were further corroborated by decreased dendritic spine density and spine head volumes that indicated impaired synapse maturation. Slice biotinylation followed by Western blot analysis of PFC-L5 tissue revealed that 5XFAD mice showed reduced synaptic AMPA receptor subunit GluA1 and decreased synaptic NMDA receptor subunit GluN2A. Consistent with this, patch-clamp recording of the evoked L23>L5 synaptic responses revealed a reduced AMPA/NMDA receptor current ratio, and an increased level of AMPAR-lacking silent synapses. These results suggest that transgenic mutant forms of APP/PS1 overexpression in 5XFAD mice leads to early developmental defects of cortical circuits, which could contribute to the age-dependent synaptic pathology and neurodegeneration later in life.
Collapse
Affiliation(s)
- Chang Chen
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Jing Wei
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Xiaokuang Ma
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Baomei Xia
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Neha Shakir
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Jessica K Zhang
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Le Zhang
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Yuehua Cui
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Deveroux Ferguson
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Shenfeng Qiu
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA.
| | - Feng Bai
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
13
|
Walters RO, Haigh CL. Organoids for modeling prion diseases. Cell Tissue Res 2023; 392:97-111. [PMID: 35088182 PMCID: PMC9329493 DOI: 10.1007/s00441-022-03589-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/19/2022] [Indexed: 11/25/2022]
Abstract
Human cerebral organoids are an exciting and novel model system emerging in the field of neurobiology. Cerebral organoids are spheres of self-organizing, neuronal lineage tissue that can be differentiated from human pluripotent stem cells and that present the possibility of on-demand human neuronal cultures that can be used for non-invasively investigating diseases affecting the brain. Compared with existing humanized cell models, they provide a more comprehensive replication of the human cerebral environment. The potential of the human cerebral organoid model is only just beginning to be elucidated, but initial studies have indicated that they could prove to be a valuable model for neurodegenerative diseases such as prion disease. The application of the cerebral organoid model to prion disease, what has been learned so far and the future potential of this model are discussed in this review.
Collapse
Affiliation(s)
- Ryan O Walters
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - Cathryn L Haigh
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA.
| |
Collapse
|
14
|
HSV-1 cellular model reveals links between aggresome formation and early step of Alzheimer's disease. Transl Psychiatry 2023; 13:86. [PMID: 36898995 PMCID: PMC10006237 DOI: 10.1038/s41398-023-02376-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
Many studies highlight the potential link between the chronic degenerative Alzheimer's disease and the infection by the herpes simplex virus type-1 (HSV-1). However, the molecular mechanisms making possible this HSV-1-dependent process remain to be understood. Using neuronal cells expressing the wild type form of amyloid precursor protein (APP) infected by HSV-1, we characterized a representative cellular model of the early stage of the sporadic form of the disease and unraveled a molecular mechanism sustaining this HSV-1- Alzheimer's disease interplay. Here, we show that HSV-1 induces caspase-dependent production of the 42 amino-acid long amyloid peptide (Aβ42) oligomers followed by their accumulation in neuronal cells. Aβ42 oligomers and activated caspase 3 (casp3A) concentrate into intracytoplasmic structures observed in Alzheimer's disease neuronal cells called aggresomes. This casp3A accumulation in aggresomes during HSV-1 infection limits the execution of apoptosis until its term, similarly to an abortosis-like event occurring in Alzheimer's disease neuronal cells patients. Indeed, this particular HSV-1 driven cellular context, representative of early stages of the disease, sustains a failed apoptosis mechanism that could explain the chronic amplification of Aβ42 production characteristic of Alzheimer's disease patients. Finally, we show that combination of flurbiprofen, a non-steroidal anti-inflammatory drug (NSAID), with caspase inhibitor reduced drastically HSV-1-induced Aβ42 oligomers production. This provided mechanistic insights supporting the conclusion of clinical trials showing that NSAIDs reduced Alzheimer's disease incidence in early stage of the disease. Therefore, from our study we propose that caspase-dependent production of Aβ42 oligomers together with the abortosis-like event represents a vicious circle in early Alzheimer's disease stages leading to a chronic amplification of Aβ42 oligomers that contributes to the establishment of degenerative disorder like Alzheimer's disease in patients infected by HSV-1. Interestingly this process could be targeted by an association of NSAID with caspase inhibitors.
Collapse
|
15
|
Rahman MM, Islam MR, Alam Tumpa MA, Shohag S, Shakil Khan Shuvo, Ferdous J, Kajol SA, Aljohani ASM, Al Abdulmonem W, Rauf A, Thiruvengadam M. Insights into the promising prospect of medicinal chemistry studies against neurodegenerative disorders. Chem Biol Interact 2023; 373:110375. [PMID: 36739931 DOI: 10.1016/j.cbi.2023.110375] [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: 11/02/2022] [Revised: 12/06/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Medicinal chemistry is an interdisciplinary field that incorporates organic chemistry, biochemistry, physical chemistry, pharmacology, informatics, molecular biology, structural biology, cell biology, and other disciplines. Additionally, it considers molecular factors such as the mode of action of the drugs, their chemical structure-activity relationship (SAR), and pharmacokinetic aspects like absorption, distribution, metabolism, elimination, and toxicity. Neurodegenerative disorders (NDs), which are defined by the breakdown of neurons over time, are affecting an increasing number of people. Oxidative stress, particularly the increased production of Reactive Oxygen Species (ROS), plays a crucial role in the growth of various disorders, as indicated by the identification of protein, lipid, and Deoxyribonucleic acid (DNA) oxidation products in vivo. Because of their inherent nature, most biological molecules are vulnerable to ROS, even if they play a role in metabolic parameters and cell signaling. Due to their high polyunsaturated fatty acid content, low antioxidant barrier, and high oxygen uptake, neurons are particularly vulnerable to oxidation by nature. As a result, excessive ROS generation in neurons looks especially harmful, and the mechanisms associated with biomolecule oxidative destruction are several and complex. This review focuses on the formation and management of ROS, as well as their chemical characteristics (both thermodynamic and kinetic), interactions, and implications in NDs.
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Mst Afroza Alam Tumpa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Sheikh Shohag
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University Buraydah, 52571, Saudi Arabia
| | - Shakil Khan Shuvo
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Jannatul Ferdous
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Saima Akter Kajol
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University Buraydah, 52571, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine Qassim University, Buraydah, Saudi Arabia
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, 23430, Khyber Pakhtunkhwa (KP), Pakistan.
| | - Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Sciences, Konkuk University, Seoul, 05029, South Korea; Department of Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India.
| |
Collapse
|
16
|
Karmirian K, Holubiec M, Goto-Silva L, Fernandez Bessone I, Vitória G, Mello B, Alloatti M, Vanderborght B, Falzone TL, Rehen S. Modeling Alzheimer's Disease Using Human Brain Organoids. Methods Mol Biol 2023; 2561:135-158. [PMID: 36399268 DOI: 10.1007/978-1-0716-2655-9_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) is the primary cause of dementia, to date. The urgent need to understand the biological and biochemical processes related to this condition, as well as the demand for reliable in vitro models for drug screening, has led to the development of novel techniques, among which stem cell methods are of utmost relevance for AD research, particularly the development of human brain organoids. Brain organoids are three-dimensional cellular aggregates derived from induced pluripotent stem cells (iPSCs) that recreate different neural cell interactions and tissue characteristics in culture. Here, we describe the protocol for the generation of brain organoids derived from AD patients and for the analysis of AD-derived pathology. AD organoids can recapitulate beta-amyloid and tau pathological features, making them a promising model for studying the molecular mechanisms underlying disease and for in vitro drug testing.
Collapse
Affiliation(s)
- Karina Karmirian
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Rio de Janeiro, Brazil
| | - Mariana Holubiec
- Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Livia Goto-Silva
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Ivan Fernandez Bessone
- Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriela Vitória
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Beatriz Mello
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Matias Alloatti
- Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Bart Vanderborght
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Tomás L Falzone
- Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigación en Biomedicina de Buenos Aires IBioBA (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Stevens Rehen
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil.
| |
Collapse
|
17
|
Ballesteros-Álvarez J, Nguyen W, Sivapatham R, Rane A, Andersen JK. Urolithin A reduces amyloid-beta load and improves cognitive deficits uncorrelated with plaque burden in a mouse model of Alzheimer's disease. GeroScience 2022; 45:1095-1113. [PMID: 36576642 PMCID: PMC9886708 DOI: 10.1007/s11357-022-00708-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/03/2022] [Indexed: 12/29/2022] Open
Abstract
In the present study, we investigated the effects of urolithin A (UA), a metabolite generated from ellagic acid via its metabolism by gut bacteria, as an autophagy activator with potential neuroprotective activity. WT and 3xTg-AD mice were administered long-term intermittent dietary supplementation with UA. UA was found to prevent deficits in spatial memory, cued fear response, and exploratory behavior in this model. It also decreased the Aβ plaque burden in areas of the hippocampus where these protein deposits are prominent in the model. Interestingly, correlation analyses demonstrate that Aβ plaque burden positively correlates with enhanced spatial memory in 3xTg-AD mice on a control diet but not in those supplemented with UA. In contrast, Aβ42 abundance in cortical and hippocampal homogenates negatively correlate with spatial memory in UA-fed mice. Our data suggest that plaque formation may be a protective mechanism against neurodegeneration and cognitive decline and that targeting the generation of proteotoxic Aβ species might be a more successful approach in halting disease progression. UA was also found to extend lifespan in normal aging mice. Mechanistically, we demonstrate that UA is able to induce autophagy and to increase Aβ clearance in neuronal cell lines. In summary, our studies reveal UA, likely via its actions as a autophagy inducer, is capable of removing Aβ from neurons and its dietary administration prevents the onset of cognitive deficits associated with pathological Aβ deposition in the 3xTg-AD mouse model as well as extending lifespan in normal aging mice.
Collapse
Affiliation(s)
| | - Wynnie Nguyen
- Buck Institute for Research on Aging, Novato, CA USA
| | | | - Anand Rane
- Buck Institute for Research on Aging, Novato, CA USA
| | | |
Collapse
|
18
|
Li L, Zhang B, Tang X, Yu Q, He A, Lu Y, Li X. A selective degeneration of cholinergic neurons mediated by NRADD in an Alzheimer's disease mouse model. CELL INSIGHT 2022; 1:100060. [PMID: 37193353 PMCID: PMC10120297 DOI: 10.1016/j.cellin.2022.100060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 05/18/2023]
Abstract
Cholinergic neurons in the basal forebrain constitute a major source of cholinergic inputs to the forebrain, modulate diverse functions including sensory processing, memory and attention, and are vulnerable to Alzheimer's disease (AD). Recently, we classified cholinergic neurons into two distinct subpopulations; calbindin D28K-expressing (D28K+) versus D28K-lacking (D28K-) neurons. Yet, which of these two cholinergic subpopulations are selectively degenerated in AD and the molecular mechanisms underlying this selective degeneration remain unknown. Here, we reported a discovery that D28K+ neurons are selectively degenerated and this degeneration induces anxiety-like behaviors in the early stage of AD. Neuronal type specific deletion of NRADD effectively rescues D28K+ neuronal degeneration, whereas genetic introduction of exogenous NRADD causes D28K- neuronal loss. This gain- and loss-of-function study reveals a subtype specific degeneration of cholinergic neurons in the disease progression of AD and hence warrants a novel molecular target for AD therapy.
Collapse
Affiliation(s)
- Lanfang Li
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bing Zhang
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaomei Tang
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Quntao Yu
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Aodi He
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Anatomy, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Youming Lu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xinyan Li
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Anatomy, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
19
|
Rickner HD, Jiang L, Hong R, O'Neill NK, Mojica CA, Snyder BJ, Zhang L, Shaw D, Medalla M, Wolozin B, Cheng CS. Single cell transcriptomic profiling of a neuron-astrocyte assembloid tauopathy model. Nat Commun 2022; 13:6275. [PMID: 36271092 PMCID: PMC9587045 DOI: 10.1038/s41467-022-34005-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 10/10/2022] [Indexed: 12/25/2022] Open
Abstract
The use of iPSC derived brain organoid models to study neurodegenerative disease has been hampered by a lack of systems that accurately and expeditiously recapitulate pathogenesis in the context of neuron-glial interactions. Here we report development of a system, termed AstTau, which propagates toxic human tau oligomers in iPSC derived neuron-astrocyte assembloids. The AstTau system develops much of the neuronal and astrocytic pathology observed in tauopathies including misfolded, phosphorylated, oligomeric, and fibrillar tau, strong neurodegeneration, and reactive astrogliosis. Single cell transcriptomic profiling combined with immunochemistry characterizes a model system that can more closely recapitulate late-stage changes in adult neurodegeneration. The transcriptomic studies demonstrate striking changes in neuroinflammatory and heat shock protein (HSP) chaperone systems in the disease process. Treatment with the HSP90 inhibitor PU-H71 is used to address the putative dysfunctional HSP chaperone system and produces a strong reduction of pathology and neurodegeneration, highlighting the potential of AstTau as a rapid and reproducible tool for drug discovery.
Collapse
Affiliation(s)
| | - Lulu Jiang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Rui Hong
- Program in Bioinformatics, Boston University, Boston, MA, 02215, USA
| | | | - Chromewell A Mojica
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Benjamin J Snyder
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Lushuang Zhang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Dipan Shaw
- Informatics Group, J. Craig Venter Institute, La Jolla, CA, 92037, USA
| | - Maria Medalla
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Benjamin Wolozin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, 02118, USA.
| | - Christine S Cheng
- Department of Biology, Boston University, Boston, MA, 02215, USA.
- Program in Bioinformatics, Boston University, Boston, MA, 02215, USA.
- Informatics Group, J. Craig Venter Institute, La Jolla, CA, 92037, USA.
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
20
|
Fan S, Li L, Liu L, Li H, Xian X, Li W. Ceftriaxone Suppresses Group II Metabotropic Glutamate Receptor Expression Contributing to Reversal of Recognition Memory Deficits of Amyloid Precursor Protein/Presenilin 1 AD Mice. Front Neurosci 2022; 16:905403. [PMID: 35860293 PMCID: PMC9289516 DOI: 10.3389/fnins.2022.905403] [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: 03/27/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Group II metabotropic glutamate receptors (Group II mGluRs) are the peri-synaptic receptor of glutamatergic neurons and negatively regulate glutamate release from presynaptic neurons. Glutamate in the synaptic cleft is mainly taken into astrocytes by glutamate transporter-1 (GLT-1), which is primarily expressed in astrocytes. Increasing evidence showed that inhibiting or suppressing the activation of Group II mGluRs would contribute to the improvement of learning and memory deficits in Alzheimer’s disease (AD) animal models. Ceftriaxone (Cef) has been reported to alleviate the spatial memory deficits in AD model mice by improving GLT-1-related clearance and metabolism of glutamate. Therefore, the present study further investigates the improving effect of Cef on recognition memory deficits and the involvement of Group II mGluRs in the process using the APP/PS1 AD mouse model. Novel object recognition tests showed that the Cef treatment significantly improved the recognition memory deficits of the AD mice. The Western blot and immunohistochemistry analysis showed that the Cef treatment significantly suppressed the upregulation of Group II mGluRs expression in APP/PS1 AD mice. The above suppression effect of Cef was blocked by dihydrokainic acid, an inhibitor of GLT-1 uptake activity. Furthermore, the Cef treatment significantly restored the downregulation in the downstream molecules of Group II mGluRs activation, including the expression of PKA and phosphorylated SNAP-25 in the APP/PS1 AD mice. The Cef treatment had no effect on the content of Aβ40 and Aβ42 in the hippocampus of APP/PS1 AD mice. The above results suggested that the suppression of Group II mGluRs contributed to the Cef-induced reversal of the recognition memory deficits in APP/PS1 AD mice.
Collapse
Affiliation(s)
- ShuJuan Fan
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Li Li
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - LiRong Liu
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - He Li
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - XiaoHui Xian
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- *Correspondence: XiaoHui Xian,
| | - WenBin Li
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- WenBin Li,
| |
Collapse
|
21
|
Catalpol Ameliorates Neurotoxicity in N2a/APP695swe Cells and APP/PS1 Transgenic Mice. Neurotox Res 2022; 40:961-972. [PMID: 35699892 DOI: 10.1007/s12640-022-00524-4] [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: 02/05/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 10/18/2022]
Abstract
Alzheimer's disease (AD) causes progressive decline of memory and cognitive deficits. Because of its complicated pathogenesis, the prevention and therapy of AD remain an enormous challenge. It has been reported that catalpol possessed neuroprotective effects against AD. However, the involved mechanism still needs to be intensively studied. Therefore, the effects of catalpol on N2a/APP695swe cells and APP/PS1 mice were identified in the current study. Catalpol could improve cytotoxicity according to CCK-8 assay and ameliorate cellular morphological changes in N2a/APP695swe cells. Neuronal structural damage in the hippocampal CA1 region of APP/PS1 AD mice was improved according to HE staining and immunohistochemistry of NeuN. Meanwhile, catalpol administration ameliorated cognitive deficits confirmed by behavior performance of APP/PS1 mice. Hoechst 33,342 staining and Annexin V-FITC/PI double staining demonstrated that catalpol could reduce apoptosis in N2a/APP695swe cells. Likewise, TUNEL staining also manifested that catalpol significantly reduced apoptosis in hippocampal CA1 region of APP/PS1 mice. Catalpol administration also could improve mitochondrial functions indicated by the ameliorative mitochondrial morphology, the decreased ROS generation, and the increased MMP in N2a/APP695swe cells. Subsequently, catalpol restrained oligomerization of Aβ1-42, verified by a reduced ThT fluorescence dose- and time-dependently. Additionally, both Aβ1-40 and Aβ1-42 aggregation were decreased in N2a/APP695swe cells and APP/PS1 mice administrated with catalpol confirmed by ELISA and western blot. Western blot also showed that catalpol facilitated the phosphorylation of AKT and GSK3β, and impeded the expression of BACE1 both in vivo and in vitro. Finally, a slight alteration in lactylation, 2-hydroxyisobutyrylation, and phosphorylation were found in N2a/APP695swe cells treated with catalpol. Together, these findings manifested that catalpol served a neuroprotective effect in AD and might be a novel and expecting prophylactic or curative candidate for AD or neurodegenerative diseases therapy.
Collapse
|
22
|
Chen B, Marquez-Nostra B, Belitzky E, Toyonaga T, Tong J, Huang Y, Cai Z. PET Imaging in Animal Models of Alzheimer’s Disease. Front Neurosci 2022; 16:872509. [PMID: 35685772 PMCID: PMC9171374 DOI: 10.3389/fnins.2022.872509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
The successful development and translation of PET imaging agents targeting β-amyloid plaques and hyperphosphorylated tau tangles have allowed for in vivo detection of these hallmarks of Alzheimer’s disease (AD) antemortem. Amyloid and tau PET have been incorporated into the A/T/N scheme for AD characterization and have become an integral part of ongoing clinical trials to screen patients for enrollment, prove drug action mechanisms, and monitor therapeutic effects. Meanwhile, preclinical PET imaging in animal models of AD can provide supportive information for mechanistic studies. With the recent advancement of gene editing technologies and AD animal model development, preclinical PET imaging in AD models will further facilitate our understanding of AD pathogenesis/progression and the development of novel treatments. In this study, we review the current state-of-the-art in preclinical PET imaging using animal models of AD and suggest future research directions.
Collapse
|
23
|
Tello JA, Williams HE, Eppler RM, Steinhilb ML, Khanna M. Animal Models of Neurodegenerative Disease: Recent Advances in Fly Highlight Innovative Approaches to Drug Discovery. Front Mol Neurosci 2022; 15:883358. [PMID: 35514431 PMCID: PMC9063566 DOI: 10.3389/fnmol.2022.883358] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative diseases represent a formidable challenge to global health. As advances in other areas of medicine grant healthy living into later decades of life, aging diseases such as Alzheimer's disease (AD) and other neurodegenerative disorders can diminish the quality of these additional years, owed largely to the lack of efficacious treatments and the absence of durable cures. Alzheimer's disease prevalence is predicted to more than double in the next 30 years, affecting nearly 15 million Americans, with AD-associated costs exceeding $1 billion by 2050. Delaying onset of AD and other neurodegenerative diseases is critical to improving the quality of life for patients and reducing the burden of disease on caregivers and healthcare systems. Significant progress has been made to model disease pathogenesis and identify points of therapeutic intervention. While some researchers have contributed to our understanding of the proteins and pathways that drive biological dysfunction in disease using in vitro and in vivo models, others have provided mathematical, biophysical, and computational technologies to identify potential therapeutic compounds using in silico modeling. The most exciting phase of the drug discovery process is now: by applying a target-directed approach that leverages the strengths of multiple techniques and validates lead hits using Drosophila as an animal model of disease, we are on the fast-track to identifying novel therapeutics to restore health to those impacted by neurodegenerative disease.
Collapse
Affiliation(s)
- Judith A. Tello
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
| | - Haley E. Williams
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
| | - Robert M. Eppler
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States
| | - Michelle L. Steinhilb
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States
| | - May Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
- Department of Molecular Pathobiology, New York University, New York, NY, United States
| |
Collapse
|
24
|
Gajera CR, Fernandez R, Postupna N, Montine KS, Keene CD, Bendall SC, Montine TJ. Mass Synaptometry: Applying Mass Cytometry to Single Synapse Analysis. Methods Mol Biol 2022; 2417:69-88. [PMID: 35099792 PMCID: PMC8820390 DOI: 10.1007/978-1-0716-1916-2_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Synaptic degeneration is one of the earliest and phenotypically most significant features associated with numerous neurodegenerative conditions, including Alzheimer's and Parkinson's diseases. Synaptic changes are also known to be important in neurocognitive disorders such as schizophrenia and autism spectrum disorders. Several labs, including ours, have demonstrated that conventional (fluorescence-based) flow cytometry of individual synaptosomes is a robust and reproducible method. However, the repertoire of probes needed to assess comprehensively the type of synapse, pathologic proteins (including protein products of risk genes discovered in GWAS), and markers of stress and injury far exceeds what is achievable with conventional flow cytometry. We recently developed a method that applies CyTOF (Cytometry by Time-Of-Flight mass spectrometry) to high-dimensional analysis of individual human synaptosomes, overcoming many of the multiplexing limitations of conventional flow cytometry. We call this new method Mass Synaptometry. Here we describe the preparation of synaptosomes from human and mouse brain, the generation and quality control of the "SynTOF" (Synapse by Time-Of-Flight mass spectrometry) antibody panel, the staining protocol, and CyTOF parameter setup for acquisition, post-acquisition processing, and analysis.
Collapse
Affiliation(s)
- Chandresh R. Gajera
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Rosemary Fernandez
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Nadia Postupna
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Kathleen S. Montine
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - C. Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Sean C. Bendall
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Thomas J. Montine
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| |
Collapse
|
25
|
Fernandes A, Caldeira C, Cunha C, Ferreiro E, Vaz AR, Brites D. Differences in Immune-Related Genes Underlie Temporal and Regional Pathological Progression in 3xTg-AD Mice. Cells 2022; 11:cells11010137. [PMID: 35011699 PMCID: PMC8750089 DOI: 10.3390/cells11010137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 01/27/2023] Open
Abstract
The prevalence of Alzheimer’s disease (AD), the most common cause of age-associated dementia, is estimated to increase over the next decades. Evidence suggests neuro-immune signaling deregulation and risk genes beyond the amyloid-β (Aβ) deposition in AD pathology. We examined the temporal profile of inflammatory mediators and microglia deactivation/activation in the brain cortex and hippocampus of 3xTg-AD mice at 3- and 9-month-old. We found upregulated APP processing, decreased expression of CD11b, CX3CR1, MFG-E8, TNF-α, IL-1β, MHC-II and C/EBP-α and increased miR-146a in both brain regions in 3-month-old 3xTG-AD mice, suggestive of a restrictive regulation. Enhanced TNF-α, IL-1β, IL-6, iNOS, SOCS1 and Arginase 1 were only present in the hippocampus of 9-month-old animals, though elevation of HMGB1 and reduction of miR-146a and miR-124 were common features in the hippocampus and cortex regions. miR-155 increased early in the cortex and later in both regions, supporting its potential as a biomarker. Candidate downregulated target genes by cortical miR-155 included Foxo3, Runx2 and CEBPβ at 3 months and Foxo3, Runx2 and Socs1 at 9 months, which are implicated in cell survival, but also in Aβ pathology and microglia/astrocyte dysfunction. Data provide new insights across AD state trajectory, with divergent microglia phenotypes and inflammatory-associated features, and identify critical targets for drug discovery and combinatorial therapies.
Collapse
Affiliation(s)
- Adelaide Fernandes
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
- Correspondence: (A.F.); (D.B.); Tel.: +351-217946450 (D.B.)
| | - Cláudia Caldeira
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (C.C.); (C.C.)
| | - Carolina Cunha
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (C.C.); (C.C.)
- Bruno Silva-Santos Lab, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Elisabete Ferreiro
- MitoXT-Mitochondrial Toxicologu and Experimental Therapeutics Laboratory, CNC-Center for Neuroscience and Cell Biology, Universidade de Coimbra, 3004-516 Coimbra, Portugal;
- III-Institute for Interdisciplinary Research (IIIUC), Universidade de Coimbra, 3004-516 Coimbra, Portugal
| | - Ana Rita Vaz
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (C.C.); (C.C.)
| | - Dora Brites
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (C.C.); (C.C.)
- Correspondence: (A.F.); (D.B.); Tel.: +351-217946450 (D.B.)
| |
Collapse
|
26
|
Nicoll JAR, Bloom T, Clarke A, Boche D, Hilton D. BRAIN UK: Accessing NHS tissue archives for neuroscience research. Neuropathol Appl Neurobiol 2021; 48:e12766. [PMID: 34528715 DOI: 10.1111/nan.12766] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 11/29/2022]
Abstract
The purpose of BRAIN UK (the UK BRain Archive Information Network) is to make the very extensive and comprehensive National Health Service (NHS) Neuropathology archives available to the national and international neuroscience research community. The archives comprise samples of tumours and a wide range of other neurological disorders, not only from the brain but also spinal cord, peripheral nerve, muscle, eye and other organs when relevant. BRAIN UK was founded after the recognition of the importance of this large tissue resource, which was not previously readily accessible for research use. BRAIN UK has successfully engaged the majority of the regional clinical neuroscience centres in the United Kingdom to produce a centralised database of the extensive autopsy and biopsy archive. Together with a simple application process and its broad ethical approval, BRAIN UK offers researchers easy access to most of the national archives of neurological tissues and tumours (http://www.brain-uk.org). The range of tissues available reflects the spectrum of disease in society, including many conditions not covered by disease-specific brain banks, and also allows relatively large numbers of cases of uncommon conditions to be studied. BRAIN UK has supported 141 studies (2010-2020) that have generated 70 publications employing methodology as diverse as morphometrics, genetics, proteomics and methylomics. Tissue samples that would otherwise have been unused have supported valuable neuroscience research. The importance of this unique resource will only increase as molecular techniques applicable to human tissues continue to develop and technical advances permit large-scale high-throughput studies.
Collapse
Affiliation(s)
- James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Department of Cellular Pathology, University Hospital Southampton, Southampton, UK
| | - Tabitha Bloom
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Amelia Clarke
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - David Hilton
- Neuropathology, Derriford Hospital, Plymouth, UK
| |
Collapse
|
27
|
Smit T, Deshayes NAC, Borchelt DR, Kamphuis W, Middeldorp J, Hol EM. Reactive astrocytes as treatment targets in Alzheimer's disease-Systematic review of studies using the APPswePS1dE9 mouse model. Glia 2021; 69:1852-1881. [PMID: 33634529 PMCID: PMC8247905 DOI: 10.1002/glia.23981] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022]
Abstract
Astrocytes regulate synaptic communication and are essential for proper brain functioning. In Alzheimer's disease (AD) astrocytes become reactive, which is characterized by an increased expression of intermediate filament proteins and cellular hypertrophy. Reactive astrocytes are found in close association with amyloid-beta (Aβ) deposits. Synaptic communication and neuronal network function could be directly modulated by reactive astrocytes, potentially contributing to cognitive decline in AD. In this review, we focus on reactive astrocytes as treatment targets in AD in the APPswePS1dE9 AD mouse model, a widely used model to study amyloidosis and gliosis. We first give an overview of the model; that is, how it was generated, which cells express the transgenes, and the effect of its genetic background on Aβ pathology. Subsequently, to determine whether modifying reactive astrocytes in AD could influence pathogenesis and cognition, we review studies using this mouse model in which interventions were directly targeted at reactive astrocytes or had an indirect effect on reactive astrocytes. Overall, studies specifically targeting astrocytes to reduce astrogliosis showed beneficial effects on cognition, which indicates that targeting astrocytes should be included in developing novel therapies for AD.
Collapse
Affiliation(s)
- Tamar Smit
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
- Swammerdam Institute for Life SciencesCenter for Neuroscience, University of AmsterdamAmsterdamThe Netherlands
| | - Natasja A. C. Deshayes
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
- Swammerdam Institute for Life SciencesCenter for Neuroscience, University of AmsterdamAmsterdamThe Netherlands
| | - David R. Borchelt
- Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, Department of NeuroscienceUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Willem Kamphuis
- Netherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| | - Jinte Middeldorp
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
- Department of ImmunobiologyBiomedical Primate Research CentreRijswijkThe Netherlands
| | - Elly M. Hol
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
28
|
Oblak AL, Lin PB, Kotredes KP, Pandey RS, Garceau D, Williams HM, Uyar A, O'Rourke R, O'Rourke S, Ingraham C, Bednarczyk D, Belanger M, Cope ZA, Little GJ, Williams SPG, Ash C, Bleckert A, Ragan T, Logsdon BA, Mangravite LM, Sukoff Rizzo SJ, Territo PR, Carter GW, Howell GR, Sasner M, Lamb BT. Comprehensive Evaluation of the 5XFAD Mouse Model for Preclinical Testing Applications: A MODEL-AD Study. Front Aging Neurosci 2021; 13:713726. [PMID: 34366832 DOI: 10.3389/fnagi.2021.71372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/23/2021] [Indexed: 05/23/2023] Open
Abstract
The ability to investigate therapeutic interventions in animal models of neurodegenerative diseases depends on extensive characterization of the model(s) being used. There are numerous models that have been generated to study Alzheimer's disease (AD) and the underlying pathogenesis of the disease. While transgenic models have been instrumental in understanding AD mechanisms and risk factors, they are limited in the degree of characteristics displayed in comparison with AD in humans, and the full spectrum of AD effects has yet to be recapitulated in a single mouse model. The Model Organism Development and Evaluation for Late-Onset Alzheimer's Disease (MODEL-AD) consortium was assembled by the National Institute on Aging (NIA) to develop more robust animal models of AD with increased relevance to human disease, standardize the characterization of AD mouse models, improve preclinical testing in animals, and establish clinically relevant AD biomarkers, among other aims toward enhancing the translational value of AD models in clinical drug design and treatment development. Here we have conducted a detailed characterization of the 5XFAD mouse, including transcriptomics, electroencephalogram, in vivo imaging, biochemical characterization, and behavioral assessments. The data from this study is publicly available through the AD Knowledge Portal.
Collapse
Affiliation(s)
- Adrian L Oblak
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Peter B Lin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Ravi S Pandey
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Dylan Garceau
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | - Asli Uyar
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Rita O'Rourke
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | - Cynthia Ingraham
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Melisa Belanger
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Zackary A Cope
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Gabriela J Little
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Carl Ash
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Adam Bleckert
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | - Tim Ragan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | | | | | | | - Paul R Territo
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | | | | | - Bruce T Lamb
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
29
|
Oblak AL, Lin PB, Kotredes KP, Pandey RS, Garceau D, Williams HM, Uyar A, O'Rourke R, O'Rourke S, Ingraham C, Bednarczyk D, Belanger M, Cope ZA, Little GJ, Williams SPG, Ash C, Bleckert A, Ragan T, Logsdon BA, Mangravite LM, Sukoff Rizzo SJ, Territo PR, Carter GW, Howell GR, Sasner M, Lamb BT. Comprehensive Evaluation of the 5XFAD Mouse Model for Preclinical Testing Applications: A MODEL-AD Study. Front Aging Neurosci 2021; 13:713726. [PMID: 34366832 PMCID: PMC8346252 DOI: 10.3389/fnagi.2021.713726] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
The ability to investigate therapeutic interventions in animal models of neurodegenerative diseases depends on extensive characterization of the model(s) being used. There are numerous models that have been generated to study Alzheimer's disease (AD) and the underlying pathogenesis of the disease. While transgenic models have been instrumental in understanding AD mechanisms and risk factors, they are limited in the degree of characteristics displayed in comparison with AD in humans, and the full spectrum of AD effects has yet to be recapitulated in a single mouse model. The Model Organism Development and Evaluation for Late-Onset Alzheimer's Disease (MODEL-AD) consortium was assembled by the National Institute on Aging (NIA) to develop more robust animal models of AD with increased relevance to human disease, standardize the characterization of AD mouse models, improve preclinical testing in animals, and establish clinically relevant AD biomarkers, among other aims toward enhancing the translational value of AD models in clinical drug design and treatment development. Here we have conducted a detailed characterization of the 5XFAD mouse, including transcriptomics, electroencephalogram, in vivo imaging, biochemical characterization, and behavioral assessments. The data from this study is publicly available through the AD Knowledge Portal.
Collapse
Affiliation(s)
- Adrian L Oblak
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States.,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Peter B Lin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Ravi S Pandey
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Dylan Garceau
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | - Asli Uyar
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Rita O'Rourke
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | - Cynthia Ingraham
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Melisa Belanger
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Zackary A Cope
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Gabriela J Little
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Carl Ash
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Adam Bleckert
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | - Tim Ragan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | | | | | | | - Paul R Territo
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | | | | | - Bruce T Lamb
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
30
|
Kim H, Chung JY. Pathobiolgy and Management of Alzheimer's Disease. Chonnam Med J 2021; 57:108-117. [PMID: 34123738 PMCID: PMC8167446 DOI: 10.4068/cmj.2021.57.2.108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 01/02/2023] Open
Abstract
Amyloid and tau protein abnormalities have been identified as the main causes of Alzheimer's disease but exact mechanisms remain to be revealed. Especially, amyloid beta and tau protein coupling and neuroinflammatory and neurovascular contributions to Alzheimer disease are quite mysterious. Many animal models and basic biological research are trying to solve these puzzles. Known as aging processes, autophagy, mitochondrial degeneration with generation of reactive oxygen species, and age-related epigenetic modifications are also known to be associated with development of Alzheimer's disease. Environmental factors such as bacterial and viral infections, heavy metal ions, diet, sleep, stress, and gut microbiota are also risk factors of Alzheimer's disease. Future development of preventive and therapeutic modalities may be dependent on the pathobiology of Alzheimer's disease.
Collapse
Affiliation(s)
- Hoowon Kim
- Department of Neurology, Chosun University Hospital, Gwangju, Korea
| | - Ji Yeon Chung
- Department of Neurology, Chosun University Hospital, Gwangju, Korea
| |
Collapse
|
31
|
Guo L, Ravindran N, Shamsher E, Hill D, Cordeiro MF. Retinal Changes in Transgenic Mouse Models of Alzheimer's Disease. Curr Alzheimer Res 2021; 18:89-102. [PMID: 33855942 DOI: 10.2174/1567205018666210414113634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/09/2021] [Accepted: 04/05/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, the most common form of dementia. AD is characterised by amyloid-β (Aβ) plaques and neurofibrillary tangles (NFT) in the brain, in association with neuronal loss and synaptic failure, causing cognitive deficits. Accurate and early diagnosis is currently unavailable in lifespan, hampering early intervention of potential new treatments. Visual deficits have been well documented in AD patients, and the pathological changes identified in the brain are also believed to be found in the retina, an integral part of the central nervous system. Retinal changes can be detected by real-time non-invasive imaging, due to the transparent nature of the ocular media, potentially allowing an earlier diagnosis as well as monitoring disease progression and treatment outcome. Animal models are essential for AD research, and this review has a focus on retinal changes in various transgenic AD mouse models with retinal imaging and immunohistochemical analysis as well as therapeutic effects in those models. We also discuss the limitations of transgenic AD models in clinical translations.
Collapse
Affiliation(s)
- Li Guo
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Nivedita Ravindran
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Ehtesham Shamsher
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Daniel Hill
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - M Francesca Cordeiro
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
32
|
Ceyzériat K, Zilli T, Millet P, Frisoni GB, Garibotto V, Tournier BB. Learning from the Past: A Review of Clinical Trials Targeting Amyloid, Tau and Neuroinflammation in Alzheimer's Disease. Curr Alzheimer Res 2021; 17:112-125. [PMID: 32129164 DOI: 10.2174/1567205017666200304085513] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 02/11/2020] [Accepted: 03/01/2020] [Indexed: 12/31/2022]
Abstract
Alzheimer's Disease (AD) is the most common neurodegenerative disease and cause of dementia. Characterized by amyloid plaques and neurofibrillary tangles of hyperphosphorylated Tau, AD pathology has been intensively studied during the last century. After a long series of failed trials of drugs targeting amyloid or Tau deposits, currently, hope lies in the positive results of one Phase III trial, highly debated, and on other ongoing trials. In parallel, some approaches target neuroinflammation, another central feature of AD. Therapeutic strategies are initially evaluated on animal models, in which the various drugs have shown effects on the target (decreasing amyloid, Tau and neuroinflammation) and sometimes on cognitive impairment. However, it is important to keep in mind that rodent models have a less complex brain than humans and that the pathology is generally not fully represented. Although they are indispensable tools in the drug discovery process, results obtained from animal models must be viewed with caution. In this review, we focus on the current status of disease-modifying therapies targeting amyloid, Tau and neuroinflammation with particular attention on the discrepancy between positive preclinical results on animal models and failures in clinical trials.
Collapse
Affiliation(s)
- Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland.,Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University and Geneva University Hospitals, Geneva, Switzerland.,Division of Radiation Oncology, Department of Oncology, Geneva University and Geneva University Hospitals, Geneva, Switzerland
| | - Thomas Zilli
- Division of Radiation Oncology, Department of Oncology, Geneva University and Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Giovanni B Frisoni
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University and Geneva University Hospitals, Geneva, Switzerland.,IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University and Geneva University Hospitals, Geneva, Switzerland
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
33
|
Souder DC, Dreischmeier IA, Smith AB, Wright S, Martin SA, Sagar MAK, Eliceiri KW, Salamat SM, Bendlin BB, Colman RJ, Beasley TM, Anderson RM. Rhesus monkeys as a translational model for late-onset Alzheimer's disease. Aging Cell 2021; 20:e13374. [PMID: 33951283 PMCID: PMC8208787 DOI: 10.1111/acel.13374] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/22/2021] [Accepted: 04/11/2021] [Indexed: 11/28/2022] Open
Abstract
Age is a major risk factor for late-onset Alzheimer's disease (AD) but seldom features in laboratory models of the disease. Furthermore, heterogeneity in size and density of AD plaques observed in individuals are not recapitulated in transgenic mouse models, presenting an incomplete picture. We show that the amyloid plaque microenvironment is not equivalent between rodent and primate species, and that differences in the impact of AD pathology on local metabolism and inflammation might explain established differences in neurodegeneration and functional decline. Using brain tissue from transgenic APP/PSEN1 mice, rhesus monkeys with age-related amyloid plaques, and human subjects with confirmed AD, we report altered energetics in the plaque microenvironment. Metabolic features included changes in mitochondrial distribution and enzymatic activity, and changes in redox cofactors NAD(P)H that were shared among species. A greater burden of lipofuscin was detected in the brains from monkeys and humans of advanced age compared to transgenic mice. Local inflammatory signatures indexed by astrogliosis and microglial activation were detected in each species; however, the inflamed zone was considerably larger for monkeys and humans. These data demonstrate the advantage of nonhuman primates in modeling the plaque microenvironment, and provide a new framework to investigate how AD pathology might contribute to functional loss.
Collapse
Affiliation(s)
- Dylan C. Souder
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
| | | | - Alex B. Smith
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
| | - Samantha Wright
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
| | - Stephen A. Martin
- Biology of Aging Laboratory Center for American Indian and Rural Health Equity Montana State University Bozeman MT USA
| | - Md Abdul Kader Sagar
- Department of Biomedical Engineering University of Wisconsin Madison Madison WI USA
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering University of Wisconsin Madison Madison WI USA
| | - Shahriar M. Salamat
- Department of Pathology Laboratory Medicine University of Wisconsin Madison Madison WI USA
- Neurological Surgery University of Wisconsin Madison Madison WI USA
| | | | - Ricki J. Colman
- Wisconsin National Primate Research Center University of Wisconsin Madison Madison WI USA
| | - T. Mark Beasley
- Department of Biostatistics University of Alabama Birmingham AL USA
- GRECC Birmingham/Atlanta Veterans Administration Hospital Birmingham AL USA
| | - Rozalyn M. Anderson
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
- GRECC William S. Middleton Memorial Veterans Hospital Madison WI USA
| |
Collapse
|
34
|
Sciaccaluga M, Megaro A, Bellomo G, Ruffolo G, Romoli M, Palma E, Costa C. An Unbalanced Synaptic Transmission: Cause or Consequence of the Amyloid Oligomers Neurotoxicity? Int J Mol Sci 2021; 22:ijms22115991. [PMID: 34206089 PMCID: PMC8199544 DOI: 10.3390/ijms22115991] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/18/2022] Open
Abstract
Amyloid-β (Aβ) 1-40 and 1-42 peptides are key mediators of synaptic and cognitive dysfunction in Alzheimer's disease (AD). Whereas in AD, Aβ is found to act as a pro-epileptogenic factor even before plaque formation, amyloid pathology has been detected among patients with epilepsy with increased risk of developing AD. Among Aβ aggregated species, soluble oligomers are suggested to be responsible for most of Aβ's toxic effects. Aβ oligomers exert extracellular and intracellular toxicity through different mechanisms, including interaction with membrane receptors and the formation of ion-permeable channels in cellular membranes. These damages, linked to an unbalance between excitatory and inhibitory neurotransmission, often result in neuronal hyperexcitability and neural circuit dysfunction, which in turn increase Aβ deposition and facilitate neurodegeneration, resulting in an Aβ-driven vicious loop. In this review, we summarize the most representative literature on the effects that oligomeric Aβ induces on synaptic dysfunction and network disorganization.
Collapse
Affiliation(s)
- Miriam Sciaccaluga
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
- Correspondence: (M.S.); (C.C.); Tel.: +39-0755858180 (M.S.); +39-0755784233 (C.C.)
| | - Alfredo Megaro
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
| | - Giovanni Bellomo
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, Istituto Pasteur—Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy; (G.R.); (E.P.)
- IRCCS San Raffaele Pisana, 00166 Rome, Italy
| | - Michele Romoli
- Neurology Unit, Rimini “Infermi” Hospital—AUSL Romagna, 47923 Rimini, Italy;
| | - Eleonora Palma
- Department of Physiology and Pharmacology, Istituto Pasteur—Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy; (G.R.); (E.P.)
| | - Cinzia Costa
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
- Correspondence: (M.S.); (C.C.); Tel.: +39-0755858180 (M.S.); +39-0755784233 (C.C.)
| |
Collapse
|
35
|
Luo J, Li P. Human pluripotent stem cell-derived brain organoids as in vitro models for studying neural disorders and cancer. Cell Biosci 2021; 11:99. [PMID: 34049587 PMCID: PMC8161602 DOI: 10.1186/s13578-021-00617-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
The sheer complexities of brain and resource limitation of human brain tissue greatly hamper our understanding of the brain disorders and cancers. Recently developed three-dimensional (3D) brain organoids (BOs) are self-organized and spontaneously differentiated from human pluripotent stem cells (hPSCs) in vitro, which exhibit similar features with cell type diversity, structural organization, and functional connectivity as the developing human brain. Based on these characteristics, hPSC-derived BOs (hPDBOs) provide new opportunities to recapitulate the complicated processes during brain development, neurodegenerative disorders, and brain cancers in vitro. In this review, we will provide an overview of existing BO models and summarize the applications of this technology in modeling the neural disorders and cancers. Furthermore, we will discuss the challenges associated with their use as in vitro models for disease modeling and the potential future direction.
Collapse
Affiliation(s)
- Juan Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
36
|
Wang H, Lv J, Zhao Y, Wei H, Zhang T, Yang H, Chen Z, Jiang J. Endothelial genetic deletion of CD147 induces changes in the dual function of the blood-brain barrier and is implicated in Alzheimer's disease. CNS Neurosci Ther 2021; 27:1048-1063. [PMID: 33987940 PMCID: PMC8339530 DOI: 10.1111/cns.13659] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 12/19/2022] Open
Abstract
AIMS The blood-brain barrier (BBB) is a specialized and indispensable structure in brain blood vessels that is damaged during Alzheimer's disease (AD). CD147 is expressed on the BBB and deeply engaged in the AD pathological process. In this study, we aimed to provide a better understanding of the roles of CD147 in BBB function in health and neurodegenerative disease. METHODS AND RESULTS We measured CD147 expression in mouse brains and demonstrated that CD147 is exclusively expressed in brain endothelial cells (BECs), and its expression decreases with age. After constructing endothelial-specific CD147 knockout mice, we performed RNA-sequencing on BECs isolated from mice of different ages as well as a range of database analyses. We found that endothelial CD147 is essential for the dual functions of the BBB, including barrier maintenance and transporter regulation. This study also shows that CD147 plays a pivotal role in neurodegenerative diseases, particularly in AD. CONCLUSIONS Our findings suggested that targeting CD147 in BECs may represent a novel therapeutic strategy, which promoted the design of future experimental investigations and the mechanistic understanding of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hao Wang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Jian‐Jun Lv
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Yu Zhao
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Hao‐Lin Wei
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Tian‐Jiao Zhang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Hai‐Jiao Yang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Zhi‐Nan Chen
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Jian‐Li Jiang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| |
Collapse
|
37
|
Dejakaisaya H, Kwan P, Jones NC. Astrocyte and glutamate involvement in the pathogenesis of epilepsy in Alzheimer's disease. Epilepsia 2021; 62:1485-1493. [PMID: 33971019 DOI: 10.1111/epi.16918] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) can increase the risk of epilepsy by up to 10-fold compared to healthy age-matched controls. However, the pathological mechanisms that underlie this increased risk are poorly understood. Because disruption in brain glutamate homeostasis has been implicated in both AD and epilepsy, this might play a mechanistic role in the pathogenesis of epilepsy in AD. Prior to the formation of amyloid beta (Aβ) plaques, the brain can undergo pathological changes as a result of increased production of amyloid precursor protein (APP) and Aβ oligomers. Impairments in the glutamate uptake ability of astrocytes due to astrogliosis are hypothesized to be an early event occurring before Aβ plaque formation. Astrogliosis may increase the susceptibility to epileptogenesis of the brain via accumulation of extracellular glutamate and resulting excitotoxicity. Here we hypothesize that Aβ oligomers and proinflammatory cytokines can cause astrogliosis and accumulation of extracellular glutamate, which then contribute to the pathogenesis of epilepsy in AD. In this review article, we consider the evidence supporting a potential role of dysfunction of the glutamate-glutamine cycle and the astrocyte in the pathogenesis of epilepsy in AD.
Collapse
Affiliation(s)
- Hattapark Dejakaisaya
- Department of Neuroscience, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Vic., Australia.,Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Patrick Kwan
- Department of Neuroscience, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Vic., Australia.,Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Vic., Australia
| | - Nigel C Jones
- Department of Neuroscience, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Vic., Australia.,Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
38
|
Alcantara-Gonzalez D, Chartampila E, Criscuolo C, Scharfman HE. Early changes in synaptic and intrinsic properties of dentate gyrus granule cells in a mouse model of Alzheimer's disease neuropathology and atypical effects of the cholinergic antagonist atropine. Neurobiol Dis 2021; 152:105274. [PMID: 33484828 DOI: 10.1016/j.nbd.2021.105274] [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] [Received: 10/01/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 12/19/2022] Open
Abstract
It has been reported that hyperexcitability occurs in a subset of patients with Alzheimer's disease (AD) and hyperexcitability could contribute to the disease. Several studies have suggested that the hippocampal dentate gyrus (DG) may be an important area where hyperexcitability occurs. Therefore, we tested the hypothesis that the principal DG cell type, granule cells (GCs), would exhibit changes at the single-cell level which would be consistent with hyperexcitability and might help explain it. We used the Tg2576 mouse, where it has been shown that hyperexcitability is robust at 2-3 months of age. GCs from 2 to 3-month-old Tg2576 mice were compared to age-matched wild type (WT) mice. Effects of muscarinic cholinergic antagonism were tested because previously we found that Tg2576 mice exhibited hyperexcitability in vivo that was reduced by the muscarinic cholinergic antagonist atropine, counter to the dogma that in AD one needs to boost cholinergic function. The results showed that GCs from Tg2576 mice exhibited increased frequency of spontaneous excitatory postsynaptic potentials/currents (sEPSP/Cs) and reduced frequency of spontaneous inhibitory synaptic events (sIPSCs) relative to WT, increasing the excitation:inhibition (E:I) ratio. There was an inward NMDA receptor-dependent current that we defined here as a novel synaptic current (nsC) in Tg2576 mice because it was very weak in WT mice. Intrinsic properties were distinct in Tg2576 GCs relative to WT. In summary, GCs of the Tg2576 mouse exhibit early electrophysiological alterations that are consistent with increased synaptic excitation, reduced inhibition, and muscarinic cholinergic dysregulation. The data support previous suggestions that the DG contributes to hyperexcitability and there is cholinergic dysfunction early in life in AD mouse models.
Collapse
Affiliation(s)
- David Alcantara-Gonzalez
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Elissavet Chartampila
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Chiara Criscuolo
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Helen E Scharfman
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York, NY 10016, USA; Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
39
|
Salobrar-García E, López-Cuenca I, Sánchez-Puebla L, de Hoz R, Fernández-Albarral JA, Ramírez AI, Bravo-Ferrer I, Medina V, Moro MA, Saido TC, Saito T, Salazar JJ, Ramírez JM. Retinal Thickness Changes Over Time in a Murine AD Model APP NL-F/NL-F. Front Aging Neurosci 2021; 12:625642. [PMID: 33542683 PMCID: PMC7852550 DOI: 10.3389/fnagi.2020.625642] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/15/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Alzheimer's disease (AD) may present retinal changes before brain pathology, suggesting the retina as an accessible biomarker of AD. The present work is a diachronic study using spectral domain optical coherence tomography (SD-OCT) to determine the total retinal thickness and retinal nerve fiber layer (RNFL) thickness in an APPNL−F/NL−F mouse model of AD at 6, 9, 12, 15, 17, and 20 months old compared to wild type (WT) animals. Methods: Total retinal thickness and RNFL thickness were determined. The mean total retinal thickness was analyzed following the Early Treatment Diabetic Retinopathy Study sectors. RNFL was measured in six sectors of axonal ring scans around the optic nerve. Results: In the APPNL−F/NL−F group compared to WT animals, the total retinal thickness changes observed were the following: (i) At 6-months-old, a significant thinning in the outer temporal sector was observed; (ii) at 15-months-old a significant thinning in the inner temporal and in the inner and outer inferior retinal sectors was noticed; (iii) at 17-months-old, a significant thickening in the inferior and nasal sectors was found in both inner and outer rings; and (iv) at 20-months-old, a significant thinning in the inner ring of nasal, temporal, and inferior retina and in the outer ring of superior and temporal retina was seen. In RNFL thickness, there was significant thinning in the global analysis and in nasal and inner-temporal sectors at 6 months old. Thinning was also found in the supero-temporal and nasal sectors and global value at 20 months old. Conclusions: In the APPNL−F/NL−F AD model, the retinal thickness showed thinning, possibly produced by neurodegeneration alternating with thickening caused by deposits and neuroinflammation in some areas of the retina. These changes over time are similar to those observed in the human retina and could be a biomarker for AD. The APPNL−F/NL−F AD model may help us better understand the different retinal changes during the progression of AD.
Collapse
Affiliation(s)
- Elena Salobrar-García
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain
| | - Lídia Sánchez-Puebla
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José A Fernández-Albarral
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain
| | - Ana I Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Isabel Bravo-Ferrer
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain.,Edinburgh Medical School, UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Violeta Medina
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María A Moro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, Brain Science Institute, RIKEN, Wako, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Juan J Salazar
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José M Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
40
|
Adeoye AO, Oso BJ. Investigative studies on the inhibition of amyloid-like fibrils formation by the extracts of Vernonia amygdalina Del. leaf. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-020-00535-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
41
|
Takaichi Y, Chambers JK, Takahashi K, Soeda Y, Koike R, Katsumata E, Kita C, Matsuda F, Haritani M, Takashima A, Nakayama H, Uchida K. Amyloid β and tau pathology in brains of aged pinniped species (sea lion, seal, and walrus). Acta Neuropathol Commun 2021; 9:10. [PMID: 33413691 PMCID: PMC7792306 DOI: 10.1186/s40478-020-01104-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/13/2020] [Indexed: 12/27/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by the accumulation of amyloid-β (Aβ) as senile plaques and cerebral amyloid angiopathy, and hyperphosphorylated tau (hp-tau) as neurofibrillary tangles in the brain. The AD-related pathology has been reported in several non-human animals, and most animals develop only the Aβ or tau pathology. We herein describe the Aβ and hp-tau pathology in the brains of aged pinniped species (seal, sea lion, and walrus). Molecular analyses revealed that the sequence of pinniped Aβ was identical to that of human Aβ. Histopathological examinations detected argyrophilic plaques composed of Aβ associated with dystrophic neurites in the cerebral cortex of aged pinnipeds. Astrogliosis and microglial infiltration were identified around Aβ plaques. Aβ deposits were observed in the blood vessel walls of the meninges and cerebrum. Pinniped tau protein was physiologically subjected to alternative splicing at exons 2, 3, and 10, and presented as five isoforms: two 3-repeat tau isoforms (1N3R, 2N3R) and three 4-repeat tau isoforms (0N4R, 1N4R, 2N4R); 0N3R tau isoform was absent. Histopathological examinations revealed argyrophilic fibrillar aggregates composed of hp-tau in the neuronal somata and neurites of aged pinniped brains. Few hp-tau aggregates were found in oligodendrocytes and microglia. Biochemically, hp-tau of the 3-repeat and 4-repeat isoforms was detected in brain sarkosyl-insoluble fractions. Aβ and hp-tau both predominantly accumulated in the neocortex, particularly the frontal cortex. Furthermore, the activation of GSK-3β was detected within cells containing hp-tau aggregates, and activated GSK-3β was strongly expressed in cases with severe hp-tau pathologies. The present results suggest that, in association with Aβ deposition, the activation of GSK-3β contributes to hp-tau accumulation in pinniped brains. Here, we report that pinniped species naturally accumulate Aβ and tau with aging, similar to the human AD pathology.
Collapse
|
42
|
Hamezah HS, Durani LW, Yanagisawa D, Ibrahim NF, Aizat WM, Makpol S, Wan Ngah WZ, Damanhuri HA, Tooyama I. Modulation of Proteome Profile in AβPP/PS1 Mice Hippocampus, Medial Prefrontal Cortex, and Striatum by Palm Oil Derived Tocotrienol-Rich Fraction. J Alzheimers Dis 2020; 72:229-246. [PMID: 31594216 PMCID: PMC6839455 DOI: 10.3233/jad-181171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tocotrienol-rich fraction (TRF) is a mixture of vitamin E analogs derived from palm oil. We previously demonstrated that supplementation with TRF improved cognitive function and modulated amyloid pathology in AβPP/PS1 mice brains. The current study was designed to examine proteomic profiles underlying the therapeutic effect of TRF in the brain. Proteomic analyses were performed on samples of hippocampus, medial prefrontal cortex (mPFC), and striatum using liquid chromatography coupled to Q Exactive HF Orbitrap mass spectrometry. From these analyses, we profiled a total of 5,847 proteins of which 155 proteins were differentially expressed between AβPP/PS1 and wild-type mice. TRF supplementation of these mice altered the expression of 255 proteins in the hippocampus, mPFC, and striatum. TRF also negatively modulated the expression of amyloid beta A4 protein and receptor-type tyrosine-protein phosphatase alpha protein in the hippocampus. The expression of proteins in metabolic pathways, oxidative phosphorylation, and those involved in Alzheimer’s disease were altered in the brains of AβPP/PS1 mice that received TRF supplementation.
Collapse
Affiliation(s)
- Hamizah Shahirah Hamezah
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Lina Wati Durani
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Daijiro Yanagisawa
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Nor Faeizah Ibrahim
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Wan Mohd Aizat
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Wan Zurinah Wan Ngah
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Hanafi Ahmad Damanhuri
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| |
Collapse
|
43
|
Abstract
Aβ plaques are one of the two lesions in the brain that define the neuropathological diagnosis of Alzheimer's disease. Plaques are highly diverse structures; many of them include massed, fibrillar polymers of the Aβ protein referred to as Aβ-amyloid, but some lack the defining features of amyloid. Cellular elements in 'classical' plaques include abnormal neuronal processes and reactive glial cells, but these are not present in all plaques. Plaques have been given various names since their discovery in 1892, including senile plaques, amyloid plaques, and neuritic plaques. However, with the identification in the 1980s of Aβ as the obligatory and universal component of plaques, the term 'Aβ plaques' has become a unifying term for these heterogeneous formations. Tauopathy, the second essential lesion of the Alzheimer's disease diagnostic dyad, is downstream of Aβ-proteopathy, but it is critically important for the manifestation of dementia. The etiologic link between Aβ-proteopathy and tauopathy in Alzheimer's disease remains largely undefined. Aβ plaques develop and propagate via the misfolding, self-assembly and spread of Aβ by the prion-like mechanism of seeded protein aggregation. Partially overlapping sets of risk factors and sequelae, including inflammation, genetic variations, and various environmental triggers have been linked to plaque development and idiopathic Alzheimer's disease, but no single factor has emerged as a requisite cause. The value of Aβ plaques per se as therapeutic targets is uncertain; although some plaques are sites of focal gliosis and inflammation, the complexity of inflammatory biology presents challenges to glia-directed intervention. Small, soluble, oligomeric assemblies of Aβ are enriched in the vicinity of plaques, and these probably contribute to the toxic impact of Aβ aggregation on the brain. Measures designed to reduce the production or seeded self-assembly of Aβ can impede the formation of Aβ plaques and oligomers, along with their accompanying abnormalities; given the apparent long timecourse of the emergence, maturation and proliferation of Aβ plaques in humans, such therapies are likely to be most effective when begun early in the pathogenic process, before significant damage has been done to the brain. Since their discovery in the late 19th century, Aβ plaques have, time and again, illuminated fundamental mechanisms driving neurodegeneration, and they should remain at the forefront of efforts to understand, and therefore treat, Alzheimer's disease.
Collapse
Affiliation(s)
- Lary C. Walker
- Department of Neurology and Yerkes National Primate Research Center, Emory University
| |
Collapse
|
44
|
Wan Nasri WN, Makpol S, Mazlan M, Tooyama I, Wan Ngah WZ, Damanhuri HA. Tocotrienol Rich Fraction Supplementation Modulate Brain Hippocampal Gene Expression in APPswe/PS1dE9 Alzheimer's Disease Mouse Model. J Alzheimers Dis 2020; 70:S239-S254. [PMID: 30507571 PMCID: PMC6700627 DOI: 10.3233/jad-180496] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by loss of memory and other cognitive abilities. AD is associated with aggregation of amyloid-β (Aβ) deposited in the hippocampal brain region. Our previous work has shown that tocotrienol rich fraction (TRF) supplementation was able to attenuate the blood oxidative status, improve behavior, and reduce fibrillary-type Aβ deposition in the hippocampus of an AD mouse model. In the present study, we investigate the effect of 6 months of TRF supplementation on transcriptome profile in the hippocampus of APPswe/PS1dE9 double transgenic mice. TRF supplementation can alleviate AD conditions by modulating several important genes in AD. Moreover, TRF supplementation attenuated the affected biological process and pathways that were upregulated in the AD mouse model. Our findings indicate that TRF supplementation can modulate hippocampal gene expression as well as biological processes that can potentially delay the progression of AD.
Collapse
Affiliation(s)
- Wan Nurzulaikha Wan Nasri
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Musalmah Mazlan
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
| | - Ikuo Tooyama
- Molecular Neuroscience Research Centre, Shiga University of Medical Sciences, Seta Tsukinowacho, Otsu, Shiga, Japan
| | - Wan Zurinah Wan Ngah
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Hanafi Ahmad Damanhuri
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
45
|
Li KY, Xiang XJ, Song L, Chen J, Luo B, Wen QX, Zhong BR, Zhou GF, Deng XJ, Ma YL, Hu LT, Chen GJ. Mitochondrial TXN2 attenuates amyloidogenesis via selective inhibition of BACE1 expression. J Neurochem 2020; 157:1351-1365. [PMID: 32920833 DOI: 10.1111/jnc.15184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/22/2022]
Abstract
Thioredoxin-2 (TXN2) is a mitochondrial protein and represents one of the intrinsic antioxidant enzymes. It has long been recognized that mitochondrial dysfunction and oxidative stress contribute to the pathogenesis of Alzheimer's disease (AD). We hypothesized that mitochondrial TXN2 might play a role in AD-like pathology. In this study, we found that in SH-SY5Y and HEK cells stably express full-length human amyloid-β precursor protein (HEK-APP), TXN2 silencing or over-expression selectively increased or decreased the transcription of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), respectively, without altering the protein levels of others enzymes involved in the catalytic processing of APP. As a result, β-amyloid protein (Aβ) levels were significantly decreased by TXN2. In addition, in cells treated with 3-nitropropionic acid (3-NP) that is known to increase reactive oxygen species (ROS) and promote mitochondrial dysfunction, TXN2 silencing resulted in further enhancement of BACE1 protein levels, suggesting a role of TXN2 in ROS removal. The downstream signaling might involve NFκB, as TXN2 reduced the phosphorylation of p65 and IκBα; and p65 knockdown significantly attenuated TXN2-mediated regulation of BACE1. Concomitantly, the levels of cellular ROS, apoptosis-related proteins and cell viability were altered by TXN2 silencing or over-expression. In APPswe/PS1E9 mice, an animal model of AD, the cortical and hippocampal TXN2 protein levels were decreased at 12 months but not at 6 months, suggesting an age-dependent decline. Collectively, TXN2 regulated BACE1 expression and amyloidogenesis via cellular ROS and NFκB signaling. TXN2 might serve as a potential target especially for early intervention of AD.
Collapse
Affiliation(s)
- Kun-Yi Li
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China.,Department of Neurology, the Second People's Hospital of Chengdu, Chengdu, China
| | - Xiao-Jiao Xiang
- Department of Nuclear Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Song
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Jian Chen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Biao Luo
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Qi-Xin Wen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Bi-Rou Zhong
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Gui-Feng Zhou
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xiao-Juan Deng
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yuan-Lin Ma
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Li-Tian Hu
- Department of Neurology, Nanchong Central Hospital, the Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Guo-Jun Chen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
46
|
Dare LR, Garcia A, Soares CB, Lopes L, Neves BHS, Dias DV, Mello-Carpes PB. The Reversal of Memory Deficits in an Alzheimer's Disease Model Using Physical and Cognitive Exercise. Front Behav Neurosci 2020; 14:152. [PMID: 32973471 PMCID: PMC7471627 DOI: 10.3389/fnbeh.2020.00152] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 08/03/2020] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia in the world, accounting for 50–75% of cases. Currently, there is limited treatment for AD. The current pharmacological therapy minimizes symptom progression but does not reverse brain damage. Studies focused on nonpharmacological treatment for AD have been developed to act on brain plasticity and minimize the neurotoxicity caused by the amyloid-beta (Aβ) peptide. Using a neurotoxicity model induced by Aβ in rats, the present study shows that physical (PE) and cognitive exercise (CE) reverse recognition memory deficits (with a prominent effect of long-term object recognition memory), decrease hippocampal lipid peroxidation, restore the acetylcholinesterase activity altered by Aβ neurotoxicity, and seems to reverse, at least partially, hippocampal tissue disorganization.
Collapse
Affiliation(s)
- Leticia R Dare
- Physiology Research Group, Federal University of Pampa, Uruguaiana, Brazil
| | - Alexandre Garcia
- Physiology Research Group, Federal University of Pampa, Uruguaiana, Brazil
| | - Caroline B Soares
- Physiology Research Group, Federal University of Pampa, Uruguaiana, Brazil
| | - Luiza Lopes
- Physiology Research Group, Federal University of Pampa, Uruguaiana, Brazil
| | - Ben-Hur S Neves
- Physiology Research Group, Federal University of Pampa, Uruguaiana, Brazil
| | - Daniel V Dias
- Department of Structural Biology, Federal University of Triangulo Mineiro, Uberaba, Brazil
| | | |
Collapse
|
47
|
Téglás T, Ábrahám D, Jókai M, Kondo S, Mohammadi R, Fehér J, Szabó D, Wilhelm M, Radák Z. Exercise combined with a probiotics treatment alters the microbiome, but moderately affects signalling pathways in the liver of male APP/PS1 transgenic mice. Biogerontology 2020; 21:807-815. [PMID: 32812166 PMCID: PMC7541368 DOI: 10.1007/s10522-020-09895-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022]
Abstract
It has been demonstrated that physical exercise and probiotic supplementation delay the progress of Alzheimer's Disease (AD) in male APP/PS1TG mice. However, it has also been suggested that both exercise and AD have systemic effects. We have studied the effects of exercise training and probiotic treatment on microbiome and biochemical signalling proteins in the liver. The results suggest that liver is under oxidative stress, since SOD2 levels of APP/PS1 mice were decreased when compared to a wild type of mice. Exercise training prevented this decrease. We did not find significant changes in COX4, SIRT3, PGC-1a or GLUT4 levels, while the changes in pAMPK/AMPK, pmTOR/mTOR, pS6/S6 and NRF2 levels were randomly modulated. The data suggest that exercise and probiotics-induced changes in microbiome do not strongly affect mitochondrial density or protein synthesis-related AMPK/mTOR/S6 pathways in the liver of these animals.
Collapse
Affiliation(s)
- Tímea Téglás
- Research Center for Molecular Exercise Science, University of Physical Education, Alkotas str. 44, Budapest, 1123, Hungary
| | - Dóra Ábrahám
- Research Center for Molecular Exercise Science, University of Physical Education, Alkotas str. 44, Budapest, 1123, Hungary
| | - Mátyás Jókai
- Research Center for Molecular Exercise Science, University of Physical Education, Alkotas str. 44, Budapest, 1123, Hungary
| | - Saki Kondo
- Research Center for Molecular Exercise Science, University of Physical Education, Alkotas str. 44, Budapest, 1123, Hungary.,Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, 113-8654, Japan
| | - Rezieh Mohammadi
- Research Center for Molecular Exercise Science, University of Physical Education, Alkotas str. 44, Budapest, 1123, Hungary
| | - János Fehér
- Ophthalmology Unit, NESMOS Department, Sant'Andrea Hospital, Faculty of Medicine and Psychology, "Sapienza" University of Rome, Rome, Italy
| | - Dóra Szabó
- Semmelweis University, Budapest, Hungary
| | - Marta Wilhelm
- Institute of Sport Sciences and Physical Education, Faculty of Science, University of Pécs, Pecs, 2020, Hungary.,Faculty of Sport Sciences, Waseda University, Saitama, 359-1192, Japan
| | - Zsolt Radák
- Research Center for Molecular Exercise Science, University of Physical Education, Alkotas str. 44, Budapest, 1123, Hungary. .,Institute of Sport Sciences and Physical Education, Faculty of Science, University of Pécs, Pecs, 2020, Hungary. .,Faculty of Sport Sciences, Waseda University, Saitama, 359-1192, Japan.
| |
Collapse
|
48
|
Wang C, Hao J, Liu X, Li C, Yuan X, Lee RJ, Bai T, Wang D. Isoforsythiaside Attenuates Alzheimer's Disease via Regulating Mitochondrial Function Through the PI3K/AKT Pathway. Int J Mol Sci 2020; 21:E5687. [PMID: 32784451 PMCID: PMC7460834 DOI: 10.3390/ijms21165687] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/28/2022] Open
Abstract
Improving mitochondrial dysfunction and inhibiting apoptosis has always been regarded as a treatment strategy for Alzheimer's disease (AD). Isoforsythiaside (IFY), a phenylethanoid glycoside isolated from the dried fruit of Forsythia suspensa, displays antioxidant activity. This study examined the neuroprotective effects of IFY and its underlying mechanisms. In the L-glutamate (L-Glu)-induced apoptosis of HT22 cells, IFY increased cell viability, inhibited mitochondrial apoptosis, and reduced the intracellular levels of reactive oxygen species (ROS), caspase-3, -8 and -9 after 3 h of pretreatment and 12-24 h of co-incubation. In the APPswe/PSEN1dE9 transgenic (APP/PS1) model, IFY reduced the anxiety of mice, improved their memory and cognitive ability, reduced the deposition of beta amyloid (Aβ) plaques in the brain, restrained the phosphorylation of the tau protein to form neurofibrillary tangles, inhibited the level of 4-hydroxynonenal in the brain, and improved phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway-related mitochondrial apoptosis. In Aβ1-42-induced U251 cells, IFY relieved the mitochondrial swelling, crest ruptures and increased their electron density after 3 h of pretreatment and 18-24 h of co-incubation. The improved cell viability and mitochondrial function after IFY incubation was blocked by the synthetic PI3K inhibitor LY294002. Taken together, these results suggest that IFY exerts a protective effect against AD by enhancing the expression levels of anti-apoptosis proteins and reducing the expression levels of pro-apoptosis proteins of B-cell lymphoma-2 (BCL-2) family members though activating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Chunyue Wang
- School of Life Sciences, Jilin University, Changchun 130012, China; (C.W.); (J.H.); (X.L.); (C.L.); (X.Y.)
| | - Jie Hao
- School of Life Sciences, Jilin University, Changchun 130012, China; (C.W.); (J.H.); (X.L.); (C.L.); (X.Y.)
| | - Xin Liu
- School of Life Sciences, Jilin University, Changchun 130012, China; (C.W.); (J.H.); (X.L.); (C.L.); (X.Y.)
| | - Chenliang Li
- School of Life Sciences, Jilin University, Changchun 130012, China; (C.W.); (J.H.); (X.L.); (C.L.); (X.Y.)
| | - Xuyang Yuan
- School of Life Sciences, Jilin University, Changchun 130012, China; (C.W.); (J.H.); (X.L.); (C.L.); (X.Y.)
| | - Robert J. Lee
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA;
| | - Tian Bai
- College of Computer Science and Technology, Jilin University, Changchun 130012, China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun 130012, China; (C.W.); (J.H.); (X.L.); (C.L.); (X.Y.)
| |
Collapse
|
49
|
Paroni G, Bisceglia P, Seripa D. Understanding the Amyloid Hypothesis in Alzheimer's Disease. J Alzheimers Dis 2020; 68:493-510. [PMID: 30883346 DOI: 10.3233/jad-180802] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The amyloid hypothesis (AH) is still the most accepted model to explain the pathogenesis of inherited Alzheimer's disease (IAD). However, despite the neuropathological overlapping with the non-inherited form (NIAD), AH waver in explaining NIAD. Thus, 30 years after its first statement several questions are still open, mainly regarding the role of amyloid plaques (AP) and apolipoprotein E (APOE). Accordingly, a pathogenetic model including the role of AP and APOE unifying IAD and NIAD pathogenesis is still missing. In the present understanding of the AH, we suggested that amyloid-β (Aβ) peptides production and AP formation is a physiological aging process resulting from a systemic age-related decrease in the efficiency of the proteins catabolism/clearance machinery. In this pathogenetic model Aβ peptides act as neurotoxic molecules, but only above a critical concentration [Aβ]c. A threshold mechanism triggers IAD/NIAD onset only when [Aβ]≥[Aβ]c. In this process, APOE modifies [Aβ]c threshold in an isoform-specific way. Consequently, all factors influencing Aβ anabolism, such as amyloid beta precursor protein (APP), presenilin 1 (PSEN1), and presenilin 2 (PSEN2) gene mutations, and/or Aβ catabolism/clearance could contribute to exceed the threshold [Aβ]c, being characteristic of each individual. In this model, AP formation does not depend on [Aβ]c. The present interpretation of the AH, unifying the pathogenetic theories for IAD and NIAD, will explain why AP and APOE4 may be observed in healthy aging and why they are not the cause of AD. It is clear that further studies are needed to confirm our pathogenetic model. Nevertheless, our suggestion may be useful to better understand the pathogenesis of AD.
Collapse
Affiliation(s)
- Giulia Paroni
- Research Laboratory, Complex Structure of Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Paola Bisceglia
- Research Laboratory, Complex Structure of Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Davide Seripa
- Research Laboratory, Complex Structure of Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| |
Collapse
|
50
|
Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer's disease. Mol Neurodegener 2020; 15:40. [PMID: 32677986 PMCID: PMC7364557 DOI: 10.1186/s13024-020-00391-7] [Citation(s) in RCA: 462] [Impact Index Per Article: 92.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder seen in age-dependent dementia. There is currently no effective treatment for AD, which may be attributed in part to lack of a clear underlying mechanism. Studies within the last few decades provide growing evidence for a central role of amyloid β (Aβ) and tau, as well as glial contributions to various molecular and cellular pathways in AD pathogenesis. Herein, we review recent progress with respect to Aβ- and tau-associated mechanisms, and discuss glial dysfunction in AD with emphasis on neuronal and glial receptors that mediate Aβ-induced toxicity. We also discuss other critical factors that may affect AD pathogenesis, including genetics, aging, variables related to environment, lifestyle habits, and describe the potential role of apolipoprotein E (APOE), viral and bacterial infection, sleep, and microbiota. Although we have gained much towards understanding various aspects underlying this devastating neurodegenerative disorder, greater commitment towards research in molecular mechanism, diagnostics and treatment will be needed in future AD research.
Collapse
Affiliation(s)
- Tiantian Guo
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Denghong Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Yuzhe Zeng
- Department of Orthopaedics, Orthopaedic Center of People's Liberation Army, The Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Timothy Y Huang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| | - Huaxi Xu
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| | - Yingjun Zhao
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|