1
|
De Plano LM, Saitta A, Oddo S, Caccamo A. Navigating Alzheimer's Disease Mouse Models: Age-Related Pathology and Cognitive Deficits. Biomolecules 2024; 14:1405. [PMID: 39595581 PMCID: PMC11592094 DOI: 10.3390/biom14111405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Since the mid-1990s, scientists have been generating mouse models of Alzheimer's disease to elucidate key mechanisms underlying the onset and progression of the disease and aid in developing potential therapeutic approaches. The first successful mouse model of Alzheimer's disease was reported in 1995 with the generation of the PDAPP mice, which were obtained by the overexpression of gene coding for the amyloid precursor protein (APP). Since then, scientists have used different approaches to develop other APP overexpression mice, mice overexpressing tau, or a combination of them. More recently, Saito and colleagues generated a mouse model by knocking in mutations associated with familial Alzheimer's disease into the APP gene. In this review, we will describe the most used animal models and provide a practical guide for the disease's age of onset and progression. We believe that this guide will be valuable for the planning and experimental design of studies utilizing these mouse models.
Collapse
Affiliation(s)
| | | | | | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (L.M.D.P.); (A.S.); (S.O.)
| |
Collapse
|
2
|
Wohlfert AJ, Phares J, Granholm AC. The mTOR Pathway: A Common Link Between Alzheimer's Disease and Down Syndrome. J Clin Med 2024; 13:6183. [PMID: 39458132 PMCID: PMC11508835 DOI: 10.3390/jcm13206183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Down syndrome (DS) is a chromosomal condition that causes many systemic dysregulations, leading to several possible age-related diseases including Alzheimer's disease (AD). This may be due to the triplication of the Amyloid precursor protein (APP) gene or other alterations in mechanistic pathways, such as the mTOR pathway. Impairments to upstream regulators of mTOR, such as insulin, PI3K/AKT, AMPK, and amino acid signaling, have been linked to amyloid beta plaques (Aβ) and neurofibrillary tangles (NFT), the most common AD pathologies. However, the mechanisms involved in the progression of pathology in human DS-related AD (DS-AD) are not fully investigated to date. Recent advancements in omics platforms are uncovering new insights into neurodegeneration. Genomics, spatial transcriptomics, proteomics, and metabolomics are novel methodologies that provide more data in greater detail than ever before; however, these methods have not been used to analyze the mTOR pathways in connection to DS-AD. Using these new techniques can unveil unexpected insights into pathological cellular mechanisms through an unbiased approach.
Collapse
Affiliation(s)
- Abigail J. Wohlfert
- Department of Modern Human Anatomy and Cell & Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Jeremiah Phares
- Department of Neurosurgery, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA;
| | - Ann-Charlotte Granholm
- Department of Neurosurgery, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA;
| |
Collapse
|
3
|
Dietz LT, Põld K, Györffy BA, Zharkovsky A, Sørensen JB, Pankratova S, Dmytriyeva O. A Peptide Motif Covering Splice Site B in Neuroligin-1 Binds to Aβ and Acts as a Neprilysin Inhibitor. Mol Neurobiol 2024:10.1007/s12035-024-04475-z. [PMID: 39261388 DOI: 10.1007/s12035-024-04475-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024]
Abstract
The most common cause of dementia among elderly people is Alzheimer's disease (AD). The typical symptom of AD is the decline of cognitive abilities, which is caused by loss of synaptic function. Amyloid-β (Aβ) oligomers play a significant role in the development of this synaptic dysfunction. Neuroligin-(NL)1 is a postsynaptic cell-adhesion molecule located in excitatory synapses and involved in the maintenance and modulation of synaptic contacts. A recent study has found that Aβ interacts with the soluble N-terminal fragment of NL1. The present study aimed to elucidate the role of NL1 in Aβ-induced neuropathology. Employing surface plasmon resonance and competitive ELISA, we confirmed the high-affinity binding of NL1 to the Aβ peptide. We also identified a sequence motif representing the NL1-binding site for the Aβ peptide and showed that a synthetic peptide modeled after this motif, termed neurolide, binds to the Aβ peptide with high affinity, comparable to the NL1-Aβ interaction. To assess the effect of neurolide in vivo, wild-type and 5XFAD mice were subcutaneously treated with this peptide for 10 weeks. We observed an increase in Aβ plaque formation in the cortex of neurolide-treated 5XFAD mice. Furthermore, we showed that neurolide reduces the activity of neprilysin, the predominant Aβ-degrading enzyme in the brain. Accordingly, we suggest that neurolide is the NL1-binding site for Aβ peptide, and acts as an inhibitor of neprilysin activity. Based on these data, we confirm the involvement of NL1 in the development of AD and suggest a mechanism for NL1-induced Aβ plaque formation.
Collapse
Affiliation(s)
- Lene T Dietz
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrin Põld
- Department of Pharmacology, University of Tartu, Tartu, Estonia
| | - Balázs A Györffy
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jakob B Sørensen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stanislava Pankratova
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, University Hospital of Copenhagen, Copenhagen, Denmark
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, University Hospital of Copenhagen, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2100, Copenhagen, Denmark.
| |
Collapse
|
4
|
Santoro B, Srinivas KV, Reyes I, Tian C, Masurkar AV. Cell type-specific impact of aging and Alzheimer disease on hippocampal CA1 perforant path input. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609952. [PMID: 39253428 PMCID: PMC11383042 DOI: 10.1101/2024.08.27.609952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
The perforant path (PP) carries direct inputs from entorhinal cortex to CA1 pyramidal neurons (PNs), with an impact dependent on PN position across transverse (CA1a-CA1c) and radial (superficial/deep) axes. It remains unclear how aging and Alzheimer disease (AD) affect PP input, despite its critical role in memory and early AD. Applying ex vivo recordings and two-photon microscopy in slices from mice up to 30 months old, we interrogated PP responses across PN subpopulations and compared them to Schaffer collateral and intrinsic excitability changes. We found that aging uniquely impacts PP excitatory responses, abolishing transverse and radial differences via a mechanism independent of presynaptic and membrane excitability change. This is amplified in aged 3xTg-AD mice, with further weakening of PP inputs to CA1a superficial PNs associated with distal dendritic spine loss. This demonstrates a unique feature of aging-related circuit dysfunction, with mechanistic implications related to memory impairment and synaptic vulnerability.
Collapse
|
5
|
Velázquez-Delgado C, Hernández-Ortiz E, Landa-Navarro L, Tapia-Rodríguez M, Moreno-Castilla P, Bermúdez-Rattoni F. Repeated exposure to novelty promotes resilience against the amyloid-beta effect through dopaminergic stimulation. Psychopharmacology (Berl) 2024:10.1007/s00213-024-06650-5. [PMID: 39145803 DOI: 10.1007/s00213-024-06650-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/07/2024] [Indexed: 08/16/2024]
Abstract
RATIONALE The accumulation of beta-amyloid peptide (Aβ) in the forebrain leads to cognitive dysfunction and neurodegeneration in Alzheimer's disease. Studies have shown that individuals with a consistently cognitively active lifestyle are less vulnerable to Aβ toxicity. Recent research has demonstrated that intrahippocampal Aβ can impact catecholaminergic release and spatial memory. Interestingly, exposure to novelty stimuli has been found to stimulate the release of catecholamines in the hippocampus. However, it remains uncertain whether repeated enhancing catecholamine activity can effectively alleviate cognitive impairment in individuals with Alzheimer's disease. OBJECTIVES Our primary aim was to investigate whether repeated exposure to novelty could enable cognitive resilience against Aβ. This protection could be achieved by modulating catecholaminergic activity within the hippocampus. METHODS To investigate this hypothesis, we subjected mice to three different conditions-standard housing (SH), repeated novelty (Nov), or daily social interaction (Soc) for one month. We then infused saline solution (SS) or Aβ (Aβ1-42) oligomers intrahippocampally and measured spatial memory retrieval in a Morris Water Maze (MWM). Stereological analysis and extracellular baseline dopamine levels using in vivo microdialysis were assessed in independent groups of mice. RESULTS The mice that received Aβ1-42 intrahippocampal infusions and remained in SH or Soc conditions showed impaired spatial memory retrieval. In contrast, animals subjected to the Nov protocol demonstrated remarkable resilience, showing strong spatial memory expression even after Aβ1-42 intrahippocampal infusion. The stereological analysis indicated that the Aβ1-42 infusion reduced the tyrosine hydroxylase axonal length in SH or Soc mice compared to the Nov group. Accordingly, the hippocampal extracellular dopamine levels increased significantly in the Nov groups. CONCLUSIONS These compelling results demonstrate the potential for repeated novelty exposure to strengthen the dopaminergic system and mitigate the toxic effects of Aβ1-42. They also highlight new and promising therapeutic avenues for treating and preventing AD, especially in its early stages.
Collapse
Affiliation(s)
- Cintia Velázquez-Delgado
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Eduardo Hernández-Ortiz
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Lucia Landa-Navarro
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Miguel Tapia-Rodríguez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Perla Moreno-Castilla
- Laboratory of Cognitive Resilience, Center of Aging Research (CIE), Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV, Mexico City, Mexico.
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
6
|
Radfar F, Shahbazi M, Tahmasebi Boroujeni S, Arab Ameri E, Farahmandfar M. Moderate aerobic training enhances the effectiveness of insulin therapy through hypothalamic IGF1 signaling in rat model of Alzheimer's disease. Sci Rep 2024; 14:15996. [PMID: 38987609 PMCID: PMC11237031 DOI: 10.1038/s41598-024-66637-2] [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: 01/27/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024] Open
Abstract
Alzheimer's disease (AD) is a neurological condition that is connected with a decline in a person's memory as well as their cognitive ability. One of the key topics of AD research has been the exploration of metabolic causes. We investigated the effects of treadmill exercise and intranasal insulin on learning and memory impairment and the expression of IGF1, BDNF, and GLUT4 in hypothalamus. The animals were put into 9 groups at random. In this study, we examined the impact of insulin on spatial memory in male Wistar rats and analyzed the effects of a 4-week pretreatment of moderate treadmill exercise and insulin on the mechanisms of improved hypothalamic glucose metabolism through changes in gene and protein expression of IGF1, BDNF, and GLUT4. We discovered that rat given Aβ25-35 had impaired spatial learning and memory, which was accompanied by higher levels of Aβ plaque burden in the hippocampus and lower levels of IGF1, BDNF, and GLUT4 mRNA and protein expression in the hypothalamus. Additionally, the administration of exercise training and intranasal insulin results in the enhancement of spatial learning and memory impairments, the reduction of plaque burden in the hippocampus, and the enhancement of the expression of IGF1, BDNF, and GLUT4 in the hypothalamus of rats that were treated with Aβ25-35. Our results show that the improvement of learning and spatial memory due to the improvement of metabolism and upregulation of the IGF1, BDNF, and GLUT4 pathways can be affected by pretreatment exercise and intranasal insulin.
Collapse
Affiliation(s)
- Forough Radfar
- Department of Behavioral and Cognitive Sciences in Sports, Sports and Health Sciences Faculty, University of Tehran, Tehran, 1417935837, Iran
| | - Mehdi Shahbazi
- Department of Behavioral and Cognitive Sciences in Sports, Sports and Health Sciences Faculty, University of Tehran, Tehran, 1417935837, Iran.
| | - Shahzad Tahmasebi Boroujeni
- Department of Behavioral and Cognitive Sciences in Sports, Sports and Health Sciences Faculty, University of Tehran, Tehran, 1417935837, Iran
| | - Elahe Arab Ameri
- Department of Behavioral and Cognitive Sciences in Sports, Sports and Health Sciences Faculty, University of Tehran, Tehran, 1417935837, Iran
| | - Maryam Farahmandfar
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, 14177-55469, Iran.
| |
Collapse
|
7
|
Guptarak J, Scaduto P, Tumurbaatar B, Zhang WR, Jupiter D, Taglialatela G, Fracassi A. Cognitive integrity in Non-Demented Individuals with Alzheimer's Neuropathology is associated with preservation and remodeling of dendritic spines. Alzheimers Dement 2024; 20:4677-4691. [PMID: 38829680 PMCID: PMC11247701 DOI: 10.1002/alz.13900] [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: 01/26/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/05/2024]
Abstract
INTRODUCTION Individuals referred to as Non-Demented with Alzheimer's Neuropathology (NDAN) exhibit cognitive resilience despite presenting Alzheimer's disease (AD) histopathological signs. Investigating the mechanisms behind this resilience may unveil crucial insights into AD resistance. METHODS DiI labeling technique was used to analyze dendritic spine morphology in control (CTRL), AD, and NDAN post mortem frontal cortex, particularly focusing on spine types near and far from amyloid beta (Aβ) plaques. RESULTS NDAN subjects displayed a higher spine density in regions distant from Aβ plaques versus AD patients. In distal areas from the plaques, NDAN individuals exhibited more immature spines, while AD patients had a prevalence of mature spines. Additionally, our examination of levels of Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1), a protein associated with synaptic plasticity and AD, showed significantly lower expression in AD versus NDAN and CTRL. DISCUSSION These results suggest that NDAN individuals undergo synaptic remodeling, potentially facilitated by Pin1, serving as a compensatory mechanism to preserve cognitive function despite AD pathology. HIGHLIGHTS Spine density is reduced near Aβ plaques compared to the distal area in CTRL, AD, and NDAN dendrites. NDAN shows higher spine density than AD in areas far from Aβ plaques. Far from Aβ plaques, NDAN has a higher density of immature spines, AD a higher density of mature spines. AD individuals show significantly lower levels of Pin1 compared to NDAN and CTRL.
Collapse
Affiliation(s)
- Jutatip Guptarak
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Pietro Scaduto
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Batbayar Tumurbaatar
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Wen Ru Zhang
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Daniel Jupiter
- Department of Biostatistics and Data Science, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Giulio Taglialatela
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| | - Anna Fracassi
- Department of Neurology, Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at GalvestonGalvestonTexasUSA
| |
Collapse
|
8
|
Del Moro L, Pirovano E, Rota E. Mind the Metabolic Gap: Bridging Migraine and Alzheimer's disease through Brain Insulin Resistance. Aging Dis 2024; 15:2526-2553. [PMID: 38913047 PMCID: PMC11567252 DOI: 10.14336/ad.2024.0351] [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: 03/29/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024] Open
Abstract
Brain insulin resistance has recently been described as a metabolic abnormality of brain glucose homeostasis that has been proven to downregulate insulin receptors, both in astrocytes and neurons, triggering a reduction in glucose uptake and glycogen synthesis. This condition may generate a mismatch between brain's energy reserve and expenditure, ??mainly during high metabolic demand, which could be involved in the chronification of migraine and, in the long run, at least in certain subsets of patients, in the prodromic phase of Alzheimer's disease, along a putative metabolic physiopathological continuum. Indeed, the persistent disruption of glucose homeostasis and energy supply to neurons may eventually impair protein folding, an energy-requiring process, promoting pathological changes in Alzheimer's disease, such as amyloid-β deposition and tau hyperphosphorylation. Hopefully, the "neuroenergetic hypothesis" presented herein will provide further insight on there being a conceivable metabolic bridge between chronic migraine and Alzheimer's disease, elucidating novel potential targets for the prophylactic treatment of both diseases.
Collapse
Affiliation(s)
- Lorenzo Del Moro
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy.
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Elenamaria Pirovano
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy.
| | - Eugenia Rota
- Neurology Unit, San Giacomo Hospital, Novi Ligure, ASL AL, Italy.
| |
Collapse
|
9
|
Chang J, Li Y, Shan X, Chen X, Yan X, Liu J, Zhao L. Neural stem cells promote neuroplasticity: a promising therapeutic strategy for the treatment of Alzheimer's disease. Neural Regen Res 2024; 19:619-628. [PMID: 37721293 PMCID: PMC10581561 DOI: 10.4103/1673-5374.380874] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/04/2023] [Accepted: 06/10/2023] [Indexed: 09/19/2023] Open
Abstract
Recent studies have demonstrated that neuroplasticity, such as synaptic plasticity and neurogenesis, exists throughout the normal lifespan but declines with age and is significantly impaired in individuals with Alzheimer's disease. Hence, promoting neuroplasticity may represent an effective strategy with which Alzheimer's disease can be alleviated. Due to their significant ability to self-renew, differentiate, and migrate, neural stem cells play an essential role in reversing synaptic and neuronal damage, reducing the pathology of Alzheimer's disease, including amyloid-β, tau protein, and neuroinflammation, and secreting neurotrophic factors and growth factors that are related to plasticity. These events can promote synaptic plasticity and neurogenesis to repair the microenvironment of the mammalian brain. Consequently, neural stem cells are considered to represent a potential regenerative therapy with which to improve Alzheimer's disease and other neurodegenerative diseases. In this review, we discuss how neural stem cells regulate neuroplasticity and optimize their effects to enhance their potential for treating Alzheimer's disease in the clinic.
Collapse
Affiliation(s)
- Jun Chang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yujiao Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaoqian Shan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xi Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xuhe Yan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jianwei Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
10
|
Ge X, Hu M, Zhou M, Fang X, Chen X, Geng D, Wang L, Yang X, An H, Zhang M, Lin D, Zheng M, Cui X, Wang Q, Wu Y, Zheng K, Huang XF, Yu Y. Overexpression of forebrain PTP1B leads to synaptic and cognitive impairments in obesity. Brain Behav Immun 2024; 117:456-470. [PMID: 38336024 DOI: 10.1016/j.bbi.2024.02.008] [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: 11/23/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024] Open
Abstract
Obesity has reached pandemic proportions and is a risk factor for neurodegenerative diseases, including Alzheimer's disease. Chronic inflammation is common in obese patients, but the mechanism between inflammation and cognitive impairment in obesity remains unclear. Accumulative evidence shows that protein-tyrosine phosphatase 1B (PTP1B), a neuroinflammatory and negative synaptic regulator, is involved in the pathogenesis of neurodegenerative processes. We investigated the causal role of PTP1B in obesity-induced cognitive impairment and the beneficial effect of PTP1B inhibitors in counteracting impairments of cognition, neural morphology, and signaling. We showed that obese individuals had negative relationship between serum PTP1B levels and cognitive function. Furthermore, the PTP1B level in the forebrain increased in patients with neurodegenerative diseases and obese cognitive impairment mice with the expansion of white matter, neuroinflammation and brain atrophy. PTP1B globally or forebrain-specific knockout mice on an obesogenic high-fat diet showed enhanced cognition and improved synaptic ultrastructure and proteins in the forebrain. Specifically, deleting PTP1B in leptin receptor-expressing cells improved leptin synaptic signaling and increased BDNF expression in the forebrain of obese mice. Importantly, we found that various PTP1B allosteric inhibitors (e.g., MSI-1436, well-tolerated in Phase 1 and 1b clinical trials for obesity and type II diabetes) prevented these alterations, including improving cognition, neurite outgrowth, leptin synaptic signaling and BDNF in both obese cognitive impairment mice and a neural cell model of PTP1B overexpression. These findings suggest that increased forebrain PTP1B is associated with cognitive decline in obesity, whereas inhibition of PTP1B could be a promising strategy for preventing neurodegeneration induced by obesity.
Collapse
Affiliation(s)
- Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Menglu Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoli Fang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu 221006, China
| | - Xi Chen
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, NSW 2522, Australia
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu 221006, China
| | - Li Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Huimei An
- HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Peking University, Beijing 10096, China
| | - Meng Zhang
- HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Peking University, Beijing 10096, China
| | - Danhong Lin
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoying Cui
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4113, Australia; Queensland Centre for Mental Health Research, Wacol, QLD, 4076, Australia
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China.
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Xu-Feng Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, NSW 2522, Australia.
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
11
|
Gu J, Yan C, Yin S, Wu H, Liu C, Xue A, Lei X, Zhang N, Geng F. Erythrocyte membrane-coated nanocarriers modified by TGN for Alzheimer's disease. J Control Release 2024; 366:448-459. [PMID: 38128884 DOI: 10.1016/j.jconrel.2023.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disease, and the main pathological feature was β-amyloid protein (Aβ) deposition. Recently, bioactive materials-based drug delivery system has been widely investigated for the treatment of AD. In this study, we developed a red blood cells (RBC) membrane-coated polycaprolactone (PCL) nanoparticles (NPs) loading with a therapeutic agent for AD, curcumin (Cur). A functional peptide TGNYKALHPHN (TGN) was conjugated to the surface of membrane for blood-brain barrier (BBB) transport (TGN-RBC-NPs-Cur). TGN peptide can be recognized by receptors on the BBB and has great potential for brain transport. To confirm the targeted delivery of Cur to the brain, a cell co-culturing immortalized human cerebral microvascular endothelial cells and human brain astrocytes glioblastoma (hCMEC/D3 and U-118MG) in vitro model was established. As a result, the BBB transporting ratio of TGN-RBC-NPs-FITC was 29.64% at 12 h which was approximately eight-fold than RBC-NPs-FITC. The improvement of drug accumulation in the AD lesion was confirmed by the NPs modified with the BBB-penetrating peptide in the fluorescence imaging and quantitative analysis with UPLC-MS/MS in vivo. The neuroprotective effects were evaluated with new object recognition behavioral test, in vitro AD cell model, dendritic spine stain, GFAP and IBA1 immunofluorescence stain. The spatial learning and memory abilities of the AD model mice treated with TGN-RBC-NPs-Cur were obviously enhanced compared with the AD control mice and were also better than Cur at the same dosage. These results were consistent with the values of protection index of rat adrenal pheochromocytoma cells (PC12 cells) treated by Aβ25-35. TGN-RBC-NPs-Cur increased the dendritic segments densities and restrained activation of microglia and astrocytes of AD mice, as well as reversed cognitive function of AD mice. All of the results demonstrated TGN-RBC-NPs-Cur a promising therapeutic strategy for delaying the progression of AD by designing biomimetic nanosystems to deliver drugs into the brain.
Collapse
Affiliation(s)
- Jinlian Gu
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China
| | - Chang Yan
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China
| | - Shun Yin
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China
| | - Hao Wu
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China
| | - Chi Liu
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China
| | - Ao Xue
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150004, China
| | - Xia Lei
- Wuxi Traditional Chinese Medicine Hospital, Wuxi, Jiangsu 214071, China.
| | - Ning Zhang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150004, China; Wuxi Traditional Chinese Medicine Hospital, Wuxi, Jiangsu 214071, China.
| | - Fang Geng
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China.
| |
Collapse
|
12
|
Sachdeva P, Narayanan KB, Sinha JK, Gupta S, Ghosh S, Singh KK, Bhaskar R, Almutary AG, Zothantluanga JH, Kotta KK, Nelson VK, Paiva-Santos AC, Abomughaid MM, Kamal M, Iqbal D, ALHarbi MH, ALMutairi AA, Dewanjee S, Nuli MV, Vippamakula S, Jha SK, Ojha S, Jha NK. Recent Advances in Drug Delivery Systems Targeting Insulin Signalling for the Treatment of Alzheimer's Disease. J Alzheimers Dis 2024; 98:1169-1179. [PMID: 38607755 DOI: 10.3233/jad-231181] [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] [Indexed: 04/14/2024]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by the accumulation of neurofibrillary tangles and amyloid-β plaques. Recent research has unveiled the pivotal role of insulin signaling dysfunction in the pathogenesis of AD. Insulin, once thought to be unrelated to brain function, has emerged as a crucial factor in neuronal survival, synaptic plasticity, and cognitive processes. Insulin and the downstream insulin signaling molecules are found mainly in the hippocampus and cortex. Some molecules responsible for dysfunction in insulin signaling are GSK-3β, Akt, PI3K, and IRS. Irregularities in insulin signaling or insulin resistance may arise from changes in the phosphorylation levels of key molecules, which can be influenced by both stimulation and inactivity. This, in turn, is believed to be a crucial factor contributing to the development of AD, which is characterized by oxidative stress, neuroinflammation, and other pathological hallmarks. Furthermore, this route is known to be indirectly influenced by Nrf2, NF-κB, and the caspases. This mini-review delves into the intricate relationship between insulin signaling and AD, exploring how disruptions in this pathway contribute to disease progression. Moreover, we examine recent advances in drug delivery systems designed to target insulin signaling for AD treatment. From oral insulin delivery to innovative nanoparticle approaches and intranasal administration, these strategies hold promise in mitigating the impact of insulin resistance on AD. This review consolidates current knowledge to shed light on the potential of these interventions as targeted therapeutic options for AD.
Collapse
Affiliation(s)
- Punya Sachdeva
- GloNeuro, Noida, Uttar Pradesh, India
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Kannan Badri Narayanan
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| | | | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology, Rajiv Gandhi InfoTech Park, Hinjawadi, Pune, Maharashtra, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - James H Zothantluanga
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
| | - Kranthi Kumar Kotta
- College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, Karnataka, India
| | - Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy of the University of Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, University of Coimbra, Coimbra, Portugal
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah, Saudi Arabia
| | - Mohammed Hamoud ALHarbi
- Department of Infection Control, Senior Consultant of Public Health, King Khalid Hospital, Al Majmaah, Ministry of Health, Saudi Arabia
| | - Awadh Aedh ALMutairi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| | - Saikat Dewanjee
- Department of Pharmaceutical Technology, Advanced Pharmacognosy Research Laboratory, Jadavpur University, Kolkata, India
| | - Mohana Vamsi Nuli
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Shanmugam Vippamakula
- MB School of Pharmaceutical Sciences, Mohan Babu University, A. Rangampet, Tirupati, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
- Department of Biotechnology, School of Applied and Life Sciences (SALS), Uttaranchal University, Dehradun, India
| |
Collapse
|
13
|
Farihi A, Bouhrim M, Chigr F, Elbouzidi A, Bencheikh N, Zrouri H, Nasr FA, Parvez MK, Alahdab A, Ahami AOT. Exploring Medicinal Herbs' Therapeutic Potential and Molecular Docking Analysis for Compounds as Potential Inhibitors of Human Acetylcholinesterase in Alzheimer's Disease Treatment. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1812. [PMID: 37893530 PMCID: PMC10608285 DOI: 10.3390/medicina59101812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: Alzheimer's disease (AD) stands as a pervasive neurodegenerative ailment of global concern, necessitating a relentless pursuit of remedies. This study aims to furnish a comprehensive exposition, delving into the intricate mechanistic actions of medicinal herbs and phytochemicals. Furthermore, we assess the potential of these compounds in inhibiting human acetylcholinesterase through molecular docking, presenting encouraging avenues for AD therapeutics. Materials and Methods: Our approach entailed a systematic exploration of phytochemicals like curcumin, gedunin, quercetin, resveratrol, nobiletin, fisetin, and berberine, targeting their capability as human acetylcholinesterase (AChE) inhibitors, leveraging the PubChem database. Diverse bioinformatics techniques were harnessed to scrutinize molecular docking, ADMET (absorption, distribution, metabolism, excretion, and toxicity), and adherence to Lipinski's rule of five. Results: Results notably underscored the substantial binding affinities of all ligands with specific amino acid residues within AChE. Remarkably, gedunin exhibited a superior binding affinity (-8.7 kcal/mol) compared to the reference standard. Conclusions: These outcomes accentuate the potential of these seven compounds as viable candidates for oral medication in AD treatment. Notably, both resveratrol and berberine demonstrated the capacity to traverse the blood-brain barrier (BBB), signaling their aptitude for central nervous system targeting. Consequently, these seven molecules are considered orally druggable, potentially surpassing the efficacy of the conventional drug, donepezil, in managing neurodegenerative disorders.
Collapse
Affiliation(s)
- Ayoub Farihi
- Unit of Clinic and Cognitive Neuroscience, Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra 14000, Morocco; (A.F.); (A.O.T.A.)
| | - Mohamed Bouhrim
- Bioengineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal 23000, Morocco; (M.B.); (F.C.); (N.B.)
| | - Fatiha Chigr
- Bioengineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal 23000, Morocco; (M.B.); (F.C.); (N.B.)
| | - Amine Elbouzidi
- Laboratory for Agricultural Production Improvement, Biotechnology, and Environment (LAPABE), Faculty of Science, Mohammed First University, Oujda 60000, Morocco
| | - Noureddine Bencheikh
- Bioengineering Laboratory, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal 23000, Morocco; (M.B.); (F.C.); (N.B.)
| | - Hassan Zrouri
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Boulevard Mohamed VI, B.P. 717, Oujda 60000, Morocco;
| | - Fahd A. Nasr
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (F.A.N.); (M.K.P.)
| | - Mohammad Khalid Parvez
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (F.A.N.); (M.K.P.)
| | - Ahmad Alahdab
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Street 17, 17489 Greifswald, Germany
| | - Ahmed Omar Touhami Ahami
- Unit of Clinic and Cognitive Neuroscience, Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra 14000, Morocco; (A.F.); (A.O.T.A.)
| |
Collapse
|
14
|
Hernández-Frausto M, Bilash OM, Masurkar AV, Basu J. Local and long-range GABAergic circuits in hippocampal area CA1 and their link to Alzheimer's disease. Front Neural Circuits 2023; 17:1223891. [PMID: 37841892 PMCID: PMC10570439 DOI: 10.3389/fncir.2023.1223891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/08/2023] [Indexed: 10/17/2023] Open
Abstract
GABAergic inhibitory neurons are the principal source of inhibition in the brain. Traditionally, their role in maintaining the balance of excitation-inhibition has been emphasized. Beyond homeostatic functions, recent circuit mapping and functional manipulation studies have revealed a wide range of specific roles that GABAergic circuits play in dynamically tilting excitation-inhibition coupling across spatio-temporal scales. These span from gating of compartment- and input-specific signaling, gain modulation, shaping input-output functions and synaptic plasticity, to generating signal-to-noise contrast, defining temporal windows for integration and rate codes, as well as organizing neural assemblies, and coordinating inter-regional synchrony. GABAergic circuits are thus instrumental in controlling single-neuron computations and behaviorally-linked network activity. The activity dependent modulation of sensory and mnemonic information processing by GABAergic circuits is pivotal for the formation and maintenance of episodic memories in the hippocampus. Here, we present an overview of the local and long-range GABAergic circuits that modulate the dynamics of excitation-inhibition and disinhibition in the main output area of the hippocampus CA1, which is crucial for episodic memory. Specifically, we link recent findings pertaining to GABAergic neuron molecular markers, electrophysiological properties, and synaptic wiring with their function at the circuit level. Lastly, given that area CA1 is particularly impaired during early stages of Alzheimer's disease, we emphasize how these GABAergic circuits may contribute to and be involved in the pathophysiology.
Collapse
Affiliation(s)
- Melissa Hernández-Frausto
- Neuroscience Institute, New York University Langone Health, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Olesia M. Bilash
- Neuroscience Institute, New York University Langone Health, New York, NY, United States
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Arjun V. Masurkar
- Neuroscience Institute, New York University Langone Health, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Jayeeta Basu
- Neuroscience Institute, New York University Langone Health, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
- Center for Neural Science, New York University, New York, NY, United States
| |
Collapse
|
15
|
Zhu WH, Yang XX, Gou XZ, Fu SM, Chen JH, Gao F, Shen Y, Bi DL, Tang AH. Nanoscale reorganisation of synaptic proteins in Alzheimer's disease. Neuropathol Appl Neurobiol 2023; 49:e12924. [PMID: 37461203 DOI: 10.1111/nan.12924] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 05/30/2023] [Accepted: 06/24/2023] [Indexed: 08/31/2023]
Abstract
AIMS Synaptic strength depends strongly on the subsynaptic organisation of presynaptic transmitter release and postsynaptic receptor densities, and their alterations are expected to underlie pathologies. Although synaptic dysfunctions are common pathogenic traits of Alzheimer's disease (AD), it remains unknown whether synaptic protein nano-organisation is altered in AD. Here, we systematically characterised the alterations in the subsynaptic organisation in cellular and mouse models of AD. METHODS We used immunostaining and super-resolution stochastic optical reconstruction microscopy imaging to quantitatively examine the synaptic protein nano-organisation in both Aβ1-42-treated neuronal cultures and cortical sections from a mouse model of AD, APP23 mice. RESULTS We found that Aβ1-42-treatment of cultured hippocampal neurons decreased the synaptic retention of postsynaptic scaffolds and receptors and disrupted their nanoscale alignment to presynaptic transmitter release sites. In cortical sections, we found that while GluA1 receptors in wild-type mice were organised in subsynaptic nanoclusters with high local densities, receptors in APP23 mice distributed more homogeneously within synapses. This reorganisation, together with the reduced overall receptor density, led to reduced glutamatergic synaptic transmission. Meanwhile, the transsynaptic alignment between presynaptic release-guiding RIM1/2 and postsynaptic scaffolding protein PSD-95 was reduced in APP23 mice. Importantly, these reorganisations were progressive with age and were more pronounced in synapses in close vicinity of Aβ plaques with dense cores. CONCLUSIONS Our study revealed a spatiotemporal-specific reorganisation of synaptic nanostructures in AD and identifies dense-core amyloid plaques as the major local inductor in APP23 mice.
Collapse
Affiliation(s)
- Wang-Hui Zhu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Xiao-Xu Yang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Xu-Zhuo Gou
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Shu-Mei Fu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Jia-Hui Chen
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Feng Gao
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
| | - Yong Shen
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, China
| | - Dan-Lei Bi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, China
| | - Ai-Hui Tang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Sciences and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| |
Collapse
|
16
|
Cai W, Li L, Sang S, Pan X, Zhong C. Physiological Roles of β-amyloid in Regulating Synaptic Function: Implications for AD Pathophysiology. Neurosci Bull 2023; 39:1289-1308. [PMID: 36443453 PMCID: PMC10387033 DOI: 10.1007/s12264-022-00985-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
The physiological functions of endogenous amyloid-β (Aβ), which plays important role in the pathology of Alzheimer's disease (AD), have not been paid enough attention. Here, we review the multiple physiological effects of Aβ, particularly in regulating synaptic transmission, and the possible mechanisms, in order to decipher the real characters of Aβ under both physiological and pathological conditions. Some worthy studies have shown that the deprivation of endogenous Aβ gives rise to synaptic dysfunction and cognitive deficiency, while the moderate elevation of this peptide enhances long term potentiation and leads to neuronal hyperexcitability. In this review, we provide a new view for understanding the role of Aβ in AD pathophysiology from the perspective of physiological meaning.
Collapse
Affiliation(s)
- Wenwen Cai
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Linxi Li
- Basic Medical College, Nanchang University, Nanchang, 330031, China
| | - Shaoming Sang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaoli Pan
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science & Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Sultana MA, Hia RA, Akinsiku O, Hegde V. Peripheral Mitochondrial Dysfunction: A Potential Contributor to the Development of Metabolic Disorders and Alzheimer's Disease. BIOLOGY 2023; 12:1019. [PMID: 37508448 PMCID: PMC10376519 DOI: 10.3390/biology12071019] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by loss of function and eventual death of neurons in the brain. Multiple studies have highlighted the involvement of mitochondria in the initiation and advancement of neurodegenerative diseases. Mitochondria are essential for ATP generation, bioenergetics processes, the regulation of calcium homeostasis and free radical scavenging. Disrupting any of these processes has been acknowledged as a major contributor to the pathogenesis of common neurodegenerative diseases, especially AD. Several longitudinal studies have demonstrated type 2 diabetes (T2D) as a risk factor for the origin of dementia leading towards AD. Even though emerging research indicates that anti-diabetic intervention is a promising option for AD prevention and therapy, results from clinical trials with anti-diabetic agents have not been effective in AD. Interestingly, defective mitochondrial function has also been reported to contribute towards the onset of metabolic disorders including obesity and T2D. The most prevalent consequences of mitochondrial dysfunction include the generation of inflammatory molecules and reactive oxygen species (ROS), which promote the onset and development of metabolic impairment and neurodegenerative diseases. Current evidence indicates an association of impaired peripheral mitochondrial function with primary AD pathology; however, the mechanisms are still unknown. Therefore, in this review, we discuss if mitochondrial dysfunction-mediated metabolic disorders have a potential connection with AD development, then would addressing peripheral mitochondrial dysfunction have better therapeutic outcomes in preventing metabolic disorder-associated AD pathologies.
Collapse
Affiliation(s)
| | | | | | - Vijay Hegde
- Obesity and Metabolic Health Laboratory, Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.A.S.); (R.A.H.); (O.A.)
| |
Collapse
|
18
|
Landry O, François A, Oye Mintsa Mi-Mba MF, Traversy MT, Tremblay C, Emond V, Bennett DA, Gylys KH, Buxbaum JD, Calon F. Postsynaptic Protein Shank3a Deficiency Synergizes with Alzheimer's Disease Neuropathology to Impair Cognitive Performance in the 3xTg-AD Murine Model. J Neurosci 2023; 43:4941-4954. [PMID: 37253603 PMCID: PMC10312061 DOI: 10.1523/jneurosci.1945-22.2023] [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: 08/09/2022] [Revised: 04/17/2023] [Accepted: 05/04/2023] [Indexed: 06/01/2023] Open
Abstract
Synaptic loss is intrinsically linked to Alzheimer's disease (AD) neuropathology and symptoms, but its direct impact on clinical symptoms remains elusive. The postsynaptic protein Shank3 (SH3 and multiple ankyrin repeat domains) is of particular interest, as the loss of a single allele of the SHANK3 gene is sufficient to cause profound cognitive symptoms in children. We thus sought to determine whether a SHANK3 deficiency could contribute to the emergence or worsening of AD symptoms and neuropathology. We first found a 30%-50% postmortem loss of SHANK3a associated with cognitive decline in the parietal cortex of individuals with AD. To further probe the role of SHANK3 in AD, we crossed male and female 3xTg-AD mice modelling Aβ and tau pathologies with Shank3a-deficient mice (Shank3Δex4-9). We observed synergistic deleterious effects of Shank3a deficiency and AD neuropathology on object recognition memory at 9, 12, and 18 months of age and on anxious behavior at 9 and 12 months of age in hemizygous Shank3Δex4-9-3xTg-AD mice. In addition to the expected 50% loss of Shank3a, levels of other synaptic proteins, such as PSD-95, drebrin, and homer1, remained unchanged in the parietotemporal cortex of hemizygous Shank3Δex4-9 animals. However, Shank3a deficiency increased the levels of soluble Aβ42 and human tau at 18 months of age compared with 3xTg-AD mice with normal Shank3 expression. The results of this study in human brain samples and in transgenic mice are consistent with the hypothesis that Shank3 deficiency makes a key contribution to cognitive impairment in AD.SIGNIFICANCE STATEMENT Although the loss of several synaptic proteins has been described in Alzheimer's disease (AD), it remains unclear whether their reduction contributes to clinical symptoms. The results of this study in human samples show lower levels of SHANK3a in AD brain, correlating with cognitive decline. Data gathered in a novel transgenic mouse suggest that Shank3a deficiency synergizes with AD neuropathology to induce cognitive impairment, consistent with a causal role in AD. Therefore, treatment aiming at preserving Shank3 in the aging brain may be beneficial to prevent AD.
Collapse
Affiliation(s)
- Olivier Landry
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Arnaud François
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Méryl-Farelle Oye Mintsa Mi-Mba
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Marie-Therese Traversy
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Vincent Emond
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - Karen H Gylys
- School of Nursing, University of California, Los Angeles, California 90095
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York 10029, New York
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| |
Collapse
|
19
|
Liu M, Zhu L, Guo YJ, Zhang SS, Jiang L, Zhang Y, Chao FL, Tang Y. The effects of voluntary running exercise on the astrocytes of the medial prefrontal cortex in APP/PS1 mice. J Comp Neurol 2023. [PMID: 37146123 DOI: 10.1002/cne.25485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 05/07/2023]
Abstract
Pathological changes in the medial prefrontal cortex (mPFC) and astrocytes are closely associated with Alzheimer's disease (AD). Voluntary running has been found to effectively delay AD. However, the effects of voluntary running on mPFC astrocytes in AD are unclear. A total of 40 10-month-old male amyloid precursor protein/presenilin 1 (APP/PS1) mice and 40 wild-type (WT) mice were randomly divided into control and running groups, and the running groups underwent voluntary running for 3 months. Mouse cognition was assessed by the novel object recognition (NOR), Morris water maze (MWM), and Y maze tests. The effects of voluntary running on mPFC astrocytes were investigated using immunohistochemistry, immunofluorescence, western blotting, and stereology. APP/PS1 mice performed significantly worse than WT mice in the NOR, MWM, and Y maze tests, and voluntary running improved the performance of APP/PS1 mice in these tests. The total number of mPFC astrocytes was increased, cell bodies were enlarged, and protrusion number and length were increased in AD mice compared with WT mice, but there was no difference in component 3 (C3) levels in the mPFC (total mPFC level); however, C3 and S100B levels in astrocytes were increased in AD mice. Voluntary running reduced the total number of astrocytes and S100B levels in astrocytes and increased the density of PSD95+ puncta in direct contact with astrocyte protrusions in the APP/PS1 mouse mPFC. Three months of voluntary running inhibited astrocyte hyperplasia and S100B expression in astrocytes, increased the density of synapses in contact with astrocytes, and improved cognitive function in APP/PS1 mice.
Collapse
Affiliation(s)
- Mei Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Lin Zhu
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Yi-Jing Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Shan-Shan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Army Medical University, Chongqing, P. R. China
| | - Lin Jiang
- Laboratory Teaching & Management Center, Chongqing Medical University, Chongqing, P. R. China
| | - Yi Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Feng-Lei Chao
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Yong Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
20
|
Kostes WW, Brafman DA. The Multifaceted Role of WNT Signaling in Alzheimer's Disease Onset and Age-Related Progression. Cells 2023; 12:1204. [PMID: 37190113 PMCID: PMC10136584 DOI: 10.3390/cells12081204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
The evolutionary conserved WNT signaling pathway orchestrates numerous complex biological processes during development and is critical to the maintenance of tissue integrity and homeostasis in the adult. As it relates to the central nervous system, WNT signaling plays several roles as it relates to neurogenesis, synaptic formation, memory, and learning. Thus, dysfunction of this pathway is associated with multiple diseases and disorders, including several neurodegenerative disorders. Alzheimer's disease (AD) is characterized by several pathologies, synaptic dysfunction, and cognitive decline. In this review, we will discuss the various epidemiological, clinical, and animal studies that demonstrate a precise link between aberrant WNT signaling and AD-associated pathologies. In turn, we will discuss the manner in which WNT signaling influences multiple molecular, biochemical, and cellular pathways upstream of these end-point pathologies. Finally, we will discuss how merging tools and technologies can be used to generate next generation cellular models to dissect the relationship between WNT signaling and AD.
Collapse
Affiliation(s)
| | - David A. Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
21
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
22
|
Activation of α7nAChR by PNU282987 improves cognitive impairment through inhibiting oxidative stress and neuroinflammation in D-galactose induced aging via regulating α7nAChR/Nrf2/HO-1 signaling pathway. Exp Gerontol 2023; 175:112139. [PMID: 36898594 DOI: 10.1016/j.exger.2023.112139] [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: 11/23/2022] [Revised: 02/22/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023]
Abstract
Aging is an important risk factor for neurodegenerative diseases. The activation of α7 nicotinic acetylcholine receptor (α7nAChR) is involved in inflammation and cognition, but the specific role it plays in aging remains unknown. This study aimed to investigate the anti-aging effect of the activation of α7nAChR on aging rats and BV2 cells induced by D-galactose, as well as its potential mechanism. D-galactose induced an increase in the SA-β-Gal positive cells, expression of p16 and p21 in vivo and in vitro. α7nAChR selective agonist PNU282987 decreased levels of pro-inflammatory factors, MDA, and Aβ, enhanced SOD activity and levels of anti-inflammatory factor (IL10) in vivo. PNU282987 enhanced the expression of Arg1, decreased the expression of iNOS, IL1β and TNFα in vitro. PNU282987 upregulated the levels of α7nAChR, Nrf2 and HO-1 in vivo and in vitro. The results of Morris water maze and novel object recognition tests showed that PNU282987 improved cognitive impairment in aging rats. Furthermore, α7nAChR selective inhibitor methyllycaconitine (MLA) results were opposite with PNU282987. PNU282987 improves cognitive impairment through inhibiting oxidative stress and neuroinflammation in D-galactose induced aging via regulating the α7nAChR/Nrf2/HO-1 signaling pathway. Therefore, targeting the α7nAChR may be a viable therapeutic approach for anti-inflammaging and neurodegenerative diseases.
Collapse
|
23
|
Ge X, Zheng M, Hu M, Fang X, Geng D, Liu S, Wang L, Zhang J, Guan L, Zheng P, Xie Y, Pan W, Zhou M, Zhou L, Tang R, Zheng K, Yu Y, Huang XF. Butyrate ameliorates quinolinic acid-induced cognitive decline in obesity models. J Clin Invest 2023; 133:154612. [PMID: 36787221 PMCID: PMC9927952 DOI: 10.1172/jci154612] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/20/2022] [Indexed: 02/15/2023] Open
Abstract
Obesity is a risk factor for neurodegenerative disease associated with cognitive dysfunction, including Alzheimer's disease. Low-grade inflammation is common in obesity, but the mechanism between inflammation and cognitive impairment in obesity is unclear. Accumulative evidence shows that quinolinic acid (QA), a neuroinflammatory neurotoxin, is involved in the pathogenesis of neurodegenerative processes. We investigated the role of QA in obesity-induced cognitive impairment and the beneficial effect of butyrate in counteracting impairments of cognition, neural morphology, and signaling. We show that in human obesity, there was a negative relationship between serum QA levels and cognitive function and decreased cortical gray matter. Diet-induced obese mice had increased QA levels in the cortex associated with cognitive impairment. At single-cell resolution, we confirmed that QA impaired neurons, altered the dendritic spine's intracellular signal, and reduced brain-derived neurotrophic factor (BDNF) levels. Using Caenorhabditis elegans models, QA induced dopaminergic and glutamatergic neuron lesions. Importantly, the gut microbiota metabolite butyrate was able to counteract those alterations, including cognitive impairment, neuronal spine loss, and BDNF reduction in both in vivo and in vitro studies. Finally, we show that butyrate prevented QA-induced BDNF reductions by epigenetic enhancement of H3K18ac at BDNF promoters. These findings suggest that increased QA is associated with cognitive decline in obesity and that butyrate alleviates neurodegeneration.
Collapse
Affiliation(s)
- Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Xiaoli Fang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Sha Liu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Li Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Jun Zhang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Li Guan
- The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Peng Zheng
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, New South Wales, Australia
| | - Yuanyi Xie
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, New South Wales, Australia
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Menglu Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Limian Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China
| | - Xu-Feng Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Jiangsu, China.,Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, New South Wales, Australia
| |
Collapse
|
24
|
Ramírez-Hernández E, Sánchez-Maldonado C, Patricio-Martínez A, Limón ID. Amyloid-β (25-35) induces the morphological alteration of dendritic spines and decreases NR2B and PSD-95 expression in the hippocampus. Neurosci Lett 2023; 795:137030. [PMID: 36572143 DOI: 10.1016/j.neulet.2022.137030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Research on the memory impairment caused by the Amyloid-β 25-35 (Aβ25-35) peptide in animal models has provided an understanding of the causes that occurs in Alzheimer's disease. However, it is uncertain whether this cognitive impairment occurs due to disruption of information encoding and consolidation or impaired retrieval of stored memory. The aim of this study was to determine the effect of the Aβ25-35 peptide on the morphology of dendritic spines and the changes in the expression of NR2B and PSD-95 in the hippocampus associated with learning and memory deficit. Vehicle or Aβ25-35 peptide (0.1 µg/µL) was bilaterally administered into the CA1 subfield of the rat hippocampus, then tested for spatial learning and memory in the Morris Water Maze. On Day 39, the morphological changes in the CA1 of the hippocampus and dentate gyrus were examined via Golgi-Cox stain. It was observed that the Aβ25-35 peptide administered in the CA1 region of the rat hippocampus induced changes to the morphology of dendritic spines and the expression of the NR2B subunit of the NMDA receptor co-localized with both the spatial memory and PSD-95 protein in the hippocampus of learning rats. We conclude that, in soluble form, the Aβ25-35 peptide perturbs synaptic plasticity, specifically in the formation of new synapses, thus promoting the progression of memory impairment.
Collapse
Affiliation(s)
- Eleazar Ramírez-Hernández
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma Puebla, Puebla, Puebla, Mexico; Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Claudia Sánchez-Maldonado
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma Puebla, Puebla, Puebla, Mexico
| | - Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma Puebla, Puebla, Puebla, Mexico; Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ilhiucamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma Puebla, Puebla, Puebla, Mexico.
| |
Collapse
|
25
|
Ribarič S. Detecting Early Cognitive Decline in Alzheimer's Disease with Brain Synaptic Structural and Functional Evaluation. Biomedicines 2023; 11:355. [PMID: 36830892 PMCID: PMC9952956 DOI: 10.3390/biomedicines11020355] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Early cognitive decline in patients with Alzheimer's (AD) is associated with quantifiable structural and functional connectivity changes in the brain. AD dysregulation of Aβ and tau metabolism progressively disrupt normal synaptic function, leading to loss of synapses, decreased hippocampal synaptic density and early hippocampal atrophy. Advances in brain imaging techniques in living patients have enabled the transition from clinical signs and symptoms-based AD diagnosis to biomarkers-based diagnosis, with functional brain imaging techniques, quantitative EEG, and body fluids sampling. The hippocampus has a central role in semantic and episodic memory processing. This cognitive function is critically dependent on normal intrahippocampal connections and normal hippocampal functional connectivity with many cortical regions, including the perirhinal and the entorhinal cortex, parahippocampal cortex, association regions in the temporal and parietal lobes, and prefrontal cortex. Therefore, decreased hippocampal synaptic density is reflected in the altered functional connectivity of intrinsic brain networks (aka large-scale networks), including the parietal memory, default mode, and salience networks. This narrative review discusses recent critical issues related to detecting AD-associated early cognitive decline with brain synaptic structural and functional markers in high-risk or neuropsychologically diagnosed patients with subjective cognitive impairment or mild cognitive impairment.
Collapse
Affiliation(s)
- Samo Ribarič
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
26
|
Pharmacological modulation of phosphodiesterase-7 as a novel strategy for neurodegenerative disorders. Inflammopharmacology 2022; 30:2051-2061. [PMID: 36272040 DOI: 10.1007/s10787-022-01072-1] [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: 07/01/2022] [Accepted: 09/06/2022] [Indexed: 11/05/2022]
Abstract
Neurodegenerative illness develops as a result of genetic defects that cause changes at numerous levels, including genomic products and biological processes. It entails the degradation of cyclic nucleotides, cyclic adenosine monophosphate (cAMP), and cyclic guanosine monophosphate (cGMP). PDE7 modulates intracellular cAMP signalling, which is involved in numerous essential physiological and pathological processes. For the therapy of neurodegenerative illnesses, the normalization of cyclic nucleotide signalling through PDE inhibition remains intriguing. In this article, we shall examine the role of PDEs in neurodegenerative diseases. Alzheimer's disease, Multiple sclerosis, Huntington's disease, Parkinson's disease, Stroke, and Epilepsy are related to alterations in PDE7 expression in the brain. Earlier, animal models of neurological illnesses including Alzheimer's disease, Parkinson's disease, and multiple sclerosis have had significant results to PDE7 inhibitors, i.e., VP3.15; VP1.14. In addition, modulation of CAMP/CREB/GSK/PKA signalling pathways involving PDE7 in neurodegenerative diseases has been addressed. To understand the etiology, treatment options of these disorders mediated by PDE7 and its subtypes can be the focus of future research.
Collapse
|
27
|
St-Pierre MK, Carrier M, González Ibáñez F, Šimončičová E, Wallman MJ, Vallières L, Parent M, Tremblay MÈ. Ultrastructural characterization of dark microglia during aging in a mouse model of Alzheimer's disease pathology and in human post-mortem brain samples. J Neuroinflammation 2022; 19:235. [PMID: 36167544 PMCID: PMC9513936 DOI: 10.1186/s12974-022-02595-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/12/2022] [Indexed: 11/10/2022] Open
Abstract
A diverse heterogeneity of microglial cells was previously described in Alzheimer's disease (AD) pathology, including dark microglia, a state characterized by ultrastructural markers of cellular stress. To provide novel insights into the roles of dark microglia during aging in the context of AD pathology, we performed a quantitative density and ultrastructural analysis of these cells using high-throughput scanning electron microscopy in the ventral hippocampus CA1 stratum lacunosum-moleculare of 20-month-old APP-PS1 vs C57BL/6J male mice. The density of dark microglia was significantly higher in APP-PS1 vs C57BL/6J mice, with these cells accounting for nearly half of all microglia observed near amyloid-beta (Aβ) plaques. This dark microglial state interacted more with dystrophic neurites compared to other APP-PS1 microglia and possessed glycogen granules, associated with a metabolic shift toward glycolysis, which provides the first ultrastructural evidence of their presence in microglia. Dark microglia were further observed in aging human post-mortem brain samples showing similar ultrastructural features as in mouse. Overall, our results provide a quantitative ultrastructural characterization of a microglial state associated with cellular stress (i.e., dark microglia) that is primarily restricted near Aβ plaques and dystrophic neurites. The presence of this microglial state in the aging human post-mortem brain is further revealed.
Collapse
Affiliation(s)
- Marie-Kim St-Pierre
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Department of Molecular Medicine, Université Laval, Québec City, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Micaël Carrier
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Fernando González Ibáñez
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Eva Šimončičová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.,Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Marie-Josée Wallman
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Quebec, QC, Canada.,CERVO Brain Research Center, Quebec, QC, Canada
| | - Luc Vallières
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
| | - Martin Parent
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Quebec, QC, Canada.,CERVO Brain Research Center, Quebec, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada. .,Department of Molecular Medicine, Université Laval, Québec City, QC, Canada. .,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada. .,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada. .,Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
28
|
Zhou R, Zhu L, Zeng Z, Luo R, Zhang J, Guo R, Zhang L, Zhang Q, Bi W. Targeted brain delivery of RVG29-modified rifampicin-loaded nanoparticles for Alzheimer's disease treatment and diagnosis. Bioeng Transl Med 2022; 7:e10395. [PMID: 36176608 PMCID: PMC9472014 DOI: 10.1002/btm2.10395] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/19/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disease. The main pathological features of AD are β-amyloid protein (Aβ) deposition and tau protein hyperphosphorylation. Currently, there are no effective drugs for the etiological treatment of AD. Rifampicin (RIF) is a semi-synthetic broad-spectrum antibiotic with anti-β-amyloid deposition, anti-inflammatory, anti-apoptosis, and neuroprotective effects, but its application in AD treatment has been limited for its strong hydrophobicity, high toxicity, short half-life, low bioavailability, and blood-brain barrier hindrance. We designed a novel brain-targeted and MRI-characteristic nanomedicine via loading rabies virus protein 29 (RVG29), rifampicin, and Gd on poly (l-lactide) nanoparticles (RIF@PLA-PEG-Gd/Mal-RVG29). The cytotoxicity assay demonstrated that RIF@PLA-PEG-Gd/Mal-RVG29 had favorable biocompatibility and security. Fluorescence imaging in vivo showed that PLA-PEG-Gd/Mal-RVG29 could deliver rifampicin into the brain by enhancing cellular uptake and brain targeting performance, leading to improvement of the bioavailability of rifampicin. In in vivo study, RIF@PLA-PEG-Gd/Mal-RVG29 improved the spatial learning and memory capability of APP/PS1 mice in the Morris water maze, as compared to rifampicin. Immunofluorescence, TEM, immunoblotting, and H&E staining revealed that RIF@PLA-PEG-Gd/Mal-RVG29 reduced Aβ deposition in hippocampal and cortex of APP/PS1 mice, improved the damage of synaptic ultrastructure, increased the expression level of PSD95 and SYP, as well as reduced the necrosis of neurons. These findings suggest that RIF@PLA-PEG-Gd/Mal-RVG29 may be an effective strategy for the treatment of AD.
Collapse
Affiliation(s)
- Ruiyi Zhou
- Department of NeurologyThe First Affiliated Hospital, Jinan UniversityGuangzhouPeople's Republic of China
| | - Lihong Zhu
- Department of PathophysiologyKey Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan UniversityGuangzhouPeople's Republic of China
| | - Zhaohao Zeng
- Department of NeurologyThe First Affiliated Hospital, Jinan UniversityGuangzhouPeople's Republic of China
| | - Rixin Luo
- Department of NeurologyThe First Affiliated Hospital, Jinan UniversityGuangzhouPeople's Republic of China
| | - Jiawei Zhang
- Department of PathophysiologyKey Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan UniversityGuangzhouPeople's Republic of China
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education InstitutesGuangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan UniversityGuangzhouPeople's Republic of China
| | - Lei Zhang
- Department of Cerebrovascular DiseaseThe Fifth Affiliated Hospital, Sun Yat‐sen UniversityZhuhaiPeople's Republic of China
| | - Qunying Zhang
- Department of CardiologyThe Fifth Affiliated Hospital, Sun Yat‐sen UniversityZhuhaiPeople's Republic of China
| | - Wei Bi
- Department of NeurologyThe First Affiliated Hospital, Jinan UniversityGuangzhouPeople's Republic of China
| |
Collapse
|
29
|
Aβ and Tau Regulate Microglia Metabolism via Exosomes in Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081800. [PMID: 35892700 PMCID: PMC9332859 DOI: 10.3390/biomedicines10081800] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 12/17/2022] Open
Abstract
One of the most striking hallmarks shared by various neurodegenerative diseases, including Alzheimer’s disease (AD), is microglia-mediated neuroinflammation. The main pathological features of AD are extracellular amyloid-β (Aβ) plaques and intracellular tau-containing neurofibrillary tangles in the brain. Amyloid-β (Aβ) peptide and tau protein are the primary components of the plaques and tangles. The crosstalk between microglia and neurons helps maintain brain homeostasis, and the metabolic phenotype of microglia determines its polarizing phenotype. There are currently many research and development efforts to provide disease-modifying therapies for AD treatment. The main targets are Aβ and tau, but whether there is a causal relationship between neurodegenerative proteins, including Aβ oligomer and tau oligomer, and regulation of microglia metabolism in neuroinflammation is still controversial. Currently, the accumulation of Aβ and tau by exosomes or other means of propagation is proposed as a regulator in neurological disorders, leading to metabolic disorders of microglia that can play a key role in the regulation of immune cells. In this review, we propose that the accumulation of Aβ oligomer and tau oligomer can propagate to adjacent microglia through exosomes and change the neuroinflammatory microenvironment by microglia metabolic reprogramming. Clarifying the relationship between harmful proteins and microglia metabolism will help people to better understand the mechanism of crosstalk between neurons and microglia, and provide new ideas for the development of AD drugs.
Collapse
|
30
|
Khan FZ, Mostaid MS, Apu MNH. Molecular Signaling Pathway Targeted Therapeutic Potential of Thymoquinone in Alzheimer’s disease. Heliyon 2022; 8:e09874. [PMID: 35832342 PMCID: PMC9272348 DOI: 10.1016/j.heliyon.2022.e09874] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 04/07/2022] [Accepted: 06/30/2022] [Indexed: 11/29/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease with rapid progression. Black cumin (Nigella sativa) is a nutraceutical that has been investigated as a prophylactic and therapeutic agent for this disease due to its ability to prevent or retard the progression of neurodegeneration. Thymoquinone (TQ) is the main bioactive compound isolated from the seeds of black cumin. Several reports have shown that it has promising potential in the prevention and treatment of AD due to its significant antioxidative, anti-inflammatory, and antiapoptotic properties along with several other mechanisms that target the altered signaling pathways due to the disease pathogenesis. In addition, it shows anticholinesterase activity and prevents α-synuclein induced synaptic damage. The aim of this review is to summarize the potential aspects and mechanisms by which TQ imparts its action in AD.
Collapse
|
31
|
Tichko P, Kim JC, Large E, Loui P. Integrating music-based interventions with Gamma-frequency stimulation: Implications for healthy ageing. Eur J Neurosci 2022; 55:3303-3323. [PMID: 33236353 PMCID: PMC9899516 DOI: 10.1111/ejn.15059] [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: 09/12/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
In recent years, music-based interventions (MBIs) have risen in popularity as a non-invasive, sustainable form of care for treating dementia-related disorders, such as Mild Cognitive Impairment (MCI) and Alzheimer's disease (AD). Despite their clinical potential, evidence regarding the efficacy of MBIs on patient outcomes is mixed. Recently, a line of related research has begun to investigate the clinical impact of non-invasive Gamma-frequency (e.g., 40 Hz) sensory stimulation on dementia. Current work, using non-human-animal models of AD, suggests that non-invasive Gamma-frequency stimulation can remediate multiple pathophysiologies of dementia at the molecular, cellular and neural-systems scales, and, importantly, improve cognitive functioning. These findings suggest that the efficacy of MBIs could, in theory, be enhanced by incorporating Gamma-frequency stimulation into current MBI protocols. In the current review, we propose a novel clinical framework for non-invasively treating dementia-related disorders that combines previous MBIs with current approaches employing Gamma-frequency sensory stimulation. We theorize that combining MBIs with Gamma-frequency stimulation could increase the therapeutic power of MBIs by simultaneously targeting multiple biomarkers of dementia, restoring neural activity that underlies learning and memory (e.g., Gamma-frequency neural activity, Theta-Gamma coupling), and actively engaging auditory and reward networks in the brain to promote behavioural change.
Collapse
Affiliation(s)
- Parker Tichko
- Department of Music, Northeastern University, Boston, MA, USA
| | - Ji Chul Kim
- Perception, Action, Cognition (PAC) Division, Department of Psychological Sciences, University of Connecticut, Storrs, CT, USA
| | - Edward Large
- Perception, Action, Cognition (PAC) Division, Department of Psychological Sciences, University of Connecticut, Storrs, CT, USA,Center for the Ecological Study of Perception & Action (CESPA), Department of Psychological Sciences, University of Connecticut, Storrs, CT, USA,Department of Physics, University of Connecticut, Storrs, CT, USA
| | - Psyche Loui
- Department of Music, Northeastern University, Boston, MA, USA
| |
Collapse
|
32
|
Weiner S, Sauer M, Visser PJ, Tijms BM, Vorontsov E, Blennow K, Zetterberg H, Gobom J. Optimized sample preparation and data analysis for TMT proteomic analysis of cerebrospinal fluid applied to the identification of Alzheimer's disease biomarkers. Clin Proteomics 2022; 19:13. [PMID: 35568819 PMCID: PMC9107710 DOI: 10.1186/s12014-022-09354-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022] Open
Abstract
Background Cerebrospinal fluid (CSF) is an important biofluid for biomarkers of neurodegenerative diseases such as Alzheimer’s disease (AD). By employing tandem mass tag (TMT) proteomics, thousands of proteins can be quantified simultaneously in large cohorts, making it a powerful tool for biomarker discovery. However, TMT proteomics in CSF is associated with analytical challenges regarding sample preparation and data processing. In this study we address those challenges ranging from data normalization over sample preparation to sample analysis. Method Using liquid chromatography coupled to mass-spectrometry (LC–MS), we analyzed TMT multiplex samples consisting of either identical or individual CSF samples, evaluated quantification accuracy and tested the performance of different data normalization approaches. We examined MS2 and MS3 acquisition strategies regarding accuracy of quantification and performed a comparative evaluation of filter-assisted sample preparation (FASP) and an in-solution protocol. Finally, four normalization approaches (median, quantile, Total Peptide Amount, TAMPOR) were applied to the previously published European Medical Information Framework Alzheimer’s Disease Multimodal Biomarker Discovery (EMIF-AD MBD) dataset. Results The correlation of measured TMT reporter ratios with spiked-in standard peptide amounts was significantly lower for TMT multiplexes composed of individual CSF samples compared with those composed of aliquots of a single CSF pool, demonstrating that the heterogeneous CSF sample composition influences TMT quantitation. Comparison of TMT reporter normalization methods showed that the correlation could be improved by applying median- and quantile-based normalization. The slope was improved by acquiring data in MS3 mode, albeit at the expense of a 29% decrease in the number of identified proteins. FASP and in-solution sample preparation of CSF samples showed a 73% overlap in identified proteins. Finally, using optimized data normalization, we present a list of 64 biomarker candidates (clinical AD vs. controls, p < 0.01) identified in the EMIF-AD cohort. Conclusion We have evaluated several analytical aspects of TMT proteomics in CSF. The results of our study provide practical guidelines to improve the accuracy of quantification and aid in the design of sample preparation and analytical protocol. The AD biomarker list extracted from the EMIF-AD cohort can provide a valuable basis for future biomarker studies and help elucidate pathogenic mechanisms in AD. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-022-09354-0.
Collapse
Affiliation(s)
- Sophia Weiner
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden.
| | - Mathias Sauer
- Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neurosciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Betty M Tijms
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neurosciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Egor Vorontsov
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute, London, UK.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
33
|
Chronic Intermittent Hypoxia Enhances Pathological Tau Seeding, Propagation, and Accumulation and Exacerbates Alzheimer-like Memory and Synaptic Plasticity Deficits and Molecular Signatures. Biol Psychiatry 2022; 91:346-358. [PMID: 34130857 PMCID: PMC8895475 DOI: 10.1016/j.biopsych.2021.02.973] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 02/19/2021] [Accepted: 02/28/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Obstructive sleep apnea, characterized by sleep fragmentation and chronic intermittent hypoxia (CIH), is a risk factor for Alzheimer's disease (AD) progression. Recent epidemiological studies point to CIH as the best predictor of developing cognitive decline and AD in older adults with obstructive sleep apnea. However, the precise underlying mechanisms remain unknown. This study was undertaken to evaluate the effect of CIH on pathological human tau seeding, propagation, and accumulation; cognition; synaptic plasticity; neuronal network excitability; and gene expression profiles in a P301S human mutant tau mouse model of AD and related tauopathies. METHODS We exposed 4- to 4.5-month-old male P301S and wild-type mice to an 8-week CIH protocol (6-min cycle: 21% O2 to 8% O2 to 21% O2, 80 cycles per 8 hours during daytime) and assessed its effect on tau pathology and various AD-related phenotypic and molecular signatures. Age- and sex-matched P301S and wild-type mice were reared in normoxia (21% O2) as experimental controls. RESULTS CIH significantly enhanced pathological human tau seeding and spread across connected brain circuitry in P301S mice; it also increased phosphorylated tau load. CIH also exacerbated memory and synaptic plasticity deficits in P301S mice. However, CIH had no effect on seizure susceptibility and network hyperexcitability in these mice. Finally, CIH exacerbated AD-related pathogenic molecular signaling in P301S mice. CONCLUSIONS CIH-induced increase in pathologic human tau seeding and spread and exacerbation of other AD-related impairments provide new insights into the role of CIH and obstructive sleep apnea in AD pathogenesis.
Collapse
|
34
|
Tuncel H, Boellaard R, Coomans EM, Hollander-Meeuwsen MD, de Vries EFJ, Glaudemans AWJM, Feltes PK, García DV, Verfaillie SCJ, Wolters EE, Sweeney SP, Ryan JM, Ivarsson M, Lynch BA, Schober P, Scheltens P, Schuit RC, Windhorst AD, De Deyn PP, van Berckel BNM, Golla SSV. Validation and test–retest repeatability performance of parametric methods for [11C]UCB-J PET. EJNMMI Res 2022; 12:3. [PMID: 35072802 PMCID: PMC8786991 DOI: 10.1186/s13550-021-00874-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/24/2021] [Indexed: 12/02/2022] Open
Abstract
[11C]UCB-J is a PET radioligand that binds to the presynaptic vesicle glycoprotein 2A. Therefore, [11C]UCB-J PET may serve as an in vivo marker of synaptic integrity. The main objective of this study was to evaluate the quantitative accuracy and the 28-day test–retest repeatability (TRT) of various parametric quantitative methods for dynamic [11C]UCB-J studies in Alzheimer’s disease (AD) patients and healthy controls (HC). Eight HCs and seven AD patients underwent two 60-min dynamic [11C]UCB-J PET scans with arterial sampling over a 28-day interval. Several plasma-input based and reference-region based parametric methods were used to generate parametric images using metabolite corrected plasma activity as input function or white matter semi-ovale as reference region. Different parametric outcomes were compared regionally with corresponding non-linear regression (NLR) estimates. Furthermore, the 28-day TRT was assessed for all parametric methods. Spectral analysis (SA) and Logan graphical analysis showed high correlations with NLR estimates. Receptor parametric mapping (RPM) and simplified reference tissue model 2 (SRTM2) BPND, and reference Logan (RLogan) distribution volume ratio (DVR) regional estimates correlated well with plasma-input derived DVR and SRTM BPND. Among the multilinear reference tissue model (MRTM) methods, MRTM1 had the best correspondence with DVR and SRTM BPND. Among the parametric methods evaluated, spectral analysis (SA) and SRTM2 were the best plasma-input and reference tissue methods, respectively, to obtain quantitatively accurate and repeatable parametric images for dynamic [11C]UCB-J PET.
Collapse
|
35
|
Abstract
The last century was characterized by a significant scientific effort aimed at unveiling the neurobiological basis of learning and memory. Thanks to the characterization of the mechanisms regulating the long-term changes of neuronal synaptic connections, it was possible to understand how specific neural networks shape themselves during the acquisition of memory traces or complex motor tasks. In this chapter, we will summarize the mechanisms underlying the main forms of synaptic plasticity taking advantage of the studies performed in the hippocampus and in the nucleus striatum, key brain structures that play a crucial role in cognition. Moreover, we will discuss how the molecular pathways involved in the induction of physiologic synaptic long-term changes could be disrupted during neurodegenerative and neuroinflammatory disorders, highlighting the translational relevance of this intriguing research field.
Collapse
Affiliation(s)
- Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Antonio de Iure
- IRCCS San Raffaele Roma, Laboratory of Experimental Neurophysiology, Rome, Italy
| | - Barbara Picconi
- IRCCS San Raffaele Roma, Laboratory of Experimental Neurophysiology, Rome, Italy; University San Raffaele, Rome, Italy.
| |
Collapse
|
36
|
Broussard JI, Redell JB, Maynard ME, Zhao J, Moore A, Mills RW, Hood KN, Underwood E, Roysam B, Dash PK. Impaired Experience-Dependent Refinement of Place Cells in a Rat Model of Alzheimer's Disease. J Alzheimers Dis 2022; 86:1907-1916. [PMID: 35253742 PMCID: PMC9850819 DOI: 10.3233/jad-215023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Hippocampal place cells play an integral role in generating spatial maps. Impaired spatial memory is a characteristic pathology of Alzheimer's disease (AD), yet it remains unclear how AD influences the properties of hippocampal place cells. OBJECTIVE To record electrophysiological activity in hippocampal CA1 neurons in freely-moving 18-month-old male TgF344-AD and age-matched wild-type (WT) littermates to examine place cell properties. METHODS We implanted 32-channel electrode arrays into the CA1 subfield of 18-month-old male WT and TgF344-AD (n = 6/group) rats. Ten days after implantation, single unit activity in an open field arena was recorded across days. The spatial information content, in-field firing rate, and stability of each place cell was compared across groups. Pathology was assessed by immunohistochemical staining, and a deep neural network approach was used to count cell profiles. RESULTS Aged TgF344-AD rats exhibited hippocampal amyloid-β deposition, and a significant increase in Iba1 immunoreactivity and microglia cell counts. Place cells from WT and TgF344-AD rat showed equivalent spatial information, in-field firing rates, and place field stability when initially exposed to the arena. However, by day 3, the place cells in aged WT rats showed characteristic spatial tuning as evidenced by higher spatial information content, stability, and in-field firing rates, an effect not seen in TgF344-AD rats. CONCLUSION These findings support the notion that altered electrophysiological properties of place cells may contribute to the learning and memory deficits observed in AD.
Collapse
Affiliation(s)
- John I. Broussard
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas 77030,To whom correspondence should be addressed: JI Broussard, Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, 6431 Fannin, St., Suite 7.011, Houston, TX 77030, Phone: (713) 500-5545,
| | - John B. Redell
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas 77030
| | - Mark E. Maynard
- Department of Electrical and Computer Engineering, Cullen College of Engineering, University of Houston, Houston, TX, 77204, USA
| | - Jing Zhao
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas 77030
| | - Anthony Moore
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas 77030
| | - Rachel W. Mills
- Department of Electrical and Computer Engineering, Cullen College of Engineering, University of Houston, Houston, TX, 77204, USA
| | - Kimberly N. Hood
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas 77030
| | - Erica Underwood
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas 77030
| | - Badrinath Roysam
- Department of Electrical and Computer Engineering, Cullen College of Engineering, University of Houston, Houston, TX, 77204, USA
| | - Pramod K. Dash
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, Texas 77030
| |
Collapse
|
37
|
Hashemi-Firouzi N, Shahidi S, Soleimani Asl S. Chronic stimulation of the serotonergic 5-HT4 receptor modulates amyloid-beta-related impairments in synaptic plasticity and memory deficits in male rats. Brain Res 2021; 1773:147701. [PMID: 34695393 DOI: 10.1016/j.brainres.2021.147701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by memory decline and impaired hippocampal synaptic plasticity. The serotonin 5-HT4 receptor is involved in learning and memory processes. This study explored the effects of chronic stimulation of 5-HT4R on cognition, memory, long-term potentiation (LTP), paired-pulse ratio (PPR), and neuronal apoptosis in a rat model of amyloid-beta (Aβ)-induced AD. Thirty-five male Wistar rats were randomly divided into three groups as follows: the sham, Aβ, and Aβ + BIMU8 groups. Aβ (6 µg/µl) was administrated by intracerebroventricular (icv) injection. The animals were treated with BIMU8 (1 μg/μL, ICV) as a 5-HT4R agonist for 30 days. Memory and behavioral changes were assessed by the passive avoidance learning, novel object recognition, open field, and elevated plus maze tests. Hippocampal synaptic plasticity was evaluated in the dentate gyrus (DG) in response to the stimulation applied to the perforant pathway. Furthermore, neuronal apoptosis was measured in the hippocampus. Data were analyzed by SPSS version 19 using one-way ANOVA, followed by Tukey's post hoc test. Aβ induced memory deficits and neuronal loss and inhibited LTP induction. Aβ also increased the normalized PPR. BIMU8 enhanced the slope of the field excitatory postsynaptic potential in LTP and improved cognition behavior. Paired-pulse inhibition or facilitation was not affected by LTP induction in Aβ animals receiving the BIMU8. It can be concluded that the stimulation of the 5-HT4 receptor modulated the Aβ-induced cognition and memory deficits, probably via a decrease in the hippocampal apoptotic neurons and an improvement in the hippocampal synaptic functions without involving its inhibitory interneurons.
Collapse
Affiliation(s)
- Nasrin Hashemi-Firouzi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Sara Soleimani Asl
- Anatomy Department, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
38
|
Ko CY, Xu JH, Lo YM, Tu RS, Wu JSB, Huang WC, Shen SC. Alleviative Effect of Alpha-Lipoic Acid on Cognitive Impairment in High-Fat Diet and Streptozotocin-Induced Type 2 Diabetic Rats. Front Aging Neurosci 2021; 13:774477. [PMID: 34867302 PMCID: PMC8633445 DOI: 10.3389/fnagi.2021.774477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/21/2021] [Indexed: 02/01/2023] Open
Abstract
Background: The intricate relationship between type 2 diabetes mellitus (T2DM) and Alzheimer’s disease (AD) suggests that insulin is involved in modulating AD-related proteins. Alpha-lipoic acid (ALA) can improve insulin resistance (IR) in diabetic rats. However, the role of ALA in alleviating the cognitive decline of T2DM is not yet clear. This study examined the ameliorative effect of ALA on cognitive impairment, cerebral IR, and synaptic plasticity abnormalities in high-fat diet (HFD) plus streptozotocin (STZ) induced diabetic rats. Methods: The HFD/STZ-induced T2DM male Wistar rats were orally administered with ALA (50, 100, or 200 mg/kg BW) once a day for 13 weeks. Abilities of cognition were measured with a passive avoidance test and Morris water maze. Specimens of blood and brain were collected for biochemical analysis after the rats were sacrificed. Western blotting was used to determine protein expressions in the hippocampus and cortex in the insulin signaling pathways, long-term potentiation (LTP), and synaptic plasticity-related protein expressions. Results: Alpha-lipoic acid improved hyperinsulinemia and the higher levels of free fatty acids of the T2DM rats. Behavioral experiments showed that the administration of ALA improved cognitive impairment in HFD/STZ-induced T2DM rats. ALA ameliorated insulin-related pathway proteins [phosphoinositide 3-kinase (PI3K), phospho-protein kinase B (pAkt)/Akt, and insulin-degrading enzyme (IDE)] and the LTP pathway, as well as synaptic plasticity proteins (calmodulin-dependent protein kinase II, cyclic AMP response element-binding protein, and postsynaptic density protein-95) of the cerebral cortex or hippocampus in HFD/STZ-induced T2DM rats. Conclusion: Our findings suggested that ALA may ameliorate cognition impairment via alleviating cerebral IR improvement and cerebral synaptic plasticity in diabetic rats.
Collapse
Affiliation(s)
- Chih-Yuan Ko
- Department of Clinical Nutrition, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.,Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.,School of Public Health, Fujian Medical University, Fuzhou, China.,Respiratory Medicine Center of Fujian Province, Quanzhou, China
| | - Jian-Hua Xu
- Department of Tumor Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | | | - Rong-Syuan Tu
- Graduate Program of Nutrition Science, National Taiwan Normal University, Taipei City, Taiwan
| | - James Swi-Bea Wu
- Graduate Institute of Food Science and Technology, National Taiwan University, Taipei City, Taiwan
| | - Wen-Chung Huang
- Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| | - Szu-Chuan Shen
- Graduate Program of Nutrition Science, National Taiwan Normal University, Taipei City, Taiwan
| |
Collapse
|
39
|
Ikeda Y, Nagase N, Tsuji A, Kitagishi Y, Matsuda S. Neuroprotection by dipeptidyl-peptidase-4 inhibitors and glucagon-like peptide-1 analogs via the modulation of AKT-signaling pathway in Alzheimer’s disease. World J Biol Chem 2021; 12:104-113. [PMID: 34904048 PMCID: PMC8637616 DOI: 10.4331/wjbc.v12.i6.104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/21/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common reason for progressive dementia in the elderly. It has been shown that disorders of the mammalian/mechanistic target of rapamycin (mTOR) signaling pathways are related to the AD. On the other hand, diabetes mellitus (DM) is a risk factor for the cognitive dysfunction. The pathogenesis of the neuronal impairment caused by diabetic hyperglycemia is intricate, which contains neuro-inflammation and/or neurodegeneration and dementia. Glucagon-like peptide-1 (GLP1) is interesting as a possible link between metabolism and brain impairment. Modulation of GLP1 activity can influence amyloid-beta peptide aggregation via the phosphoinositide-3 kinase/AKT/mTOR signaling pathway in AD. The GLP1 receptor agonists have been shown to have favorable actions on the brain such as the improvement of neurological deficit. They might also exert a beneficial effect with refining learning and memory on the cognitive impairment induced by diabetes. Recent experimental and clinical evidence indicates that dipeptidyl-peptidase-4 (DPP4) inhibitors, being currently used for DM therapy, may also be effective for AD treatment. The DPP-4 inhibitors have demonstrated neuroprotection and cognitive improvements in animal models. Although further studies for mTOR, GLP1, and DPP4 signaling pathways in humans would be intensively required, they seem to be a promising approach for innovative AD-treatments. We would like to review the characteristics of AD pathogenesis, the key roles of mTOR in AD and the preventive and/ or therapeutic suggestions of directing the mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuka Ikeda
- Food Science and Nutrition, Nara Women’s University, Nara 630-8506, Japan
| | - Nozomi Nagase
- Food Science and Nutrition, Nara Women’s University, Nara 630-8506, Japan
| | - Ai Tsuji
- Food Science and Nutrition, Nara Women’s University, Nara 630-8506, Japan
| | - Yasuko Kitagishi
- Food Science and Nutrition, Nara Women’s University, Nara 630-8506, Japan
| | - Satoru Matsuda
- Food Science and Nutrition, Nara Women’s University, Nara 630-8506, Japan
| |
Collapse
|
40
|
Jiang H, Esparza TJ, Kummer TT, Brody DL. Unbiased high-content screening reveals Aβ- and tau-independent synaptotoxic activities in human brain homogenates from Alzheimer's patients and high-pathology controls. PLoS One 2021; 16:e0259335. [PMID: 34748596 PMCID: PMC8575250 DOI: 10.1371/journal.pone.0259335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/19/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer’s disease (AD) is tightly correlated with synapse loss in vulnerable brain regions. It is assumed that specific molecular entities such as Aβ and tau cause synapse loss in AD, yet unbiased screens for synaptotoxic activities have not been performed. Here, we performed size exclusion chromatography on soluble human brain homogenates from AD cases, high pathology non-demented controls, and low pathology age-matched controls using our novel high content primary cultured neuron-based screening assay. Both presynaptic and postsynaptic toxicities were elevated in homogenates from AD cases and high pathology non-demented controls to a similar extent, with more modest synaptotoxic activities in homogenates from low pathology normal controls. Surprisingly, synaptotoxic activities were found in size fractions peaking between the 17–44 kDa size standards that did not match well with Aβ and tau immunoreactive species in these homogenates. The fractions containing previously identified high molecular weight soluble amyloid beta aggregates/”oligomers” were non-toxic in this assay. Furthermore, immunodepletion of Aβ and tau did not reduce synaptotoxic activity. This result contrasts with previous findings involving the same methods applied to 3xTg-AD mouse brain extracts. The nature of the synaptotoxic species has not been identified. Overall, our data indicates one or more potential Aβ and tau independent synaptotoxic activities in human AD brain homogenates. This result aligns well with the key role of synaptic loss in the early cognitive decline and may provide new insight into AD pathophysiology.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Thomas J. Esparza
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States of America
| | - Terrance T. Kummer
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - David L. Brody
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States of America
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
41
|
Xiao G, Chen Q, Zhang X. MicroRNA-455-5p/CPEB1 pathway mediates Aβ-related learning and memory deficits in a mouse model of Alzheimer's disease. Brain Res Bull 2021; 177:282-294. [PMID: 34678444 DOI: 10.1016/j.brainresbull.2021.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 09/12/2021] [Accepted: 10/12/2021] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD), a common neurodegenerative disease, is the main cause of dementia, with cognitive decline as the core symptom observed during diagnosis. Synaptic loss may be the main cause of early cognitive dysfunction in AD, but the detailed mechanism is still unclear. In this study, we investigated the role of abnormal miR-455-5p/CPEB1 pathway in AD mouse model. We found that miR-455-5p was upregulated, while its downstream target, cytoplasmic polyadenylation element-binding 1 (CPEB1), was downregulated in the hippocampus of APP/PS1 mice at the age of 9 m. Abnormal miR-455-5p/CPEB1 pathway mediated cognitive deficits in APP/PS1 mice through suppressing α-Amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor expressions. And miR-455-5p suppression, CPEB1 overexpression or application of a peptide disrupting the miR-455-5p/CPEB1 interaction in CA1 of APP/PS1 mice rescued AD-like phenotypes in mice, including deficits in synaptic plasticity and memory. In conclusion, our results indicated that miRNA-455-5p/CPEB1 pathway mediated synaptic and memory deficits in Alzheimer's Disease through targeting on AMPARs, providing a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Xuewei Zhang
- Department of Health Managent Center, Xiangya hospital, Central South University, Changsha, Hunan 410008, PR China; Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, PR China.
| |
Collapse
|
42
|
Monllor P, Cervera-Ferri A, Lloret MA, Esteve D, Lopez B, Leon JL, Lloret A. Electroencephalography as a Non-Invasive Biomarker of Alzheimer's Disease: A Forgotten Candidate to Substitute CSF Molecules? Int J Mol Sci 2021; 22:10889. [PMID: 34639229 PMCID: PMC8509134 DOI: 10.3390/ijms221910889] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/26/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Biomarkers for disease diagnosis and prognosis are crucial in clinical practice. They should be objective and quantifiable and respond to specific therapeutic interventions. Optimal biomarkers should reflect the underlying process (pathological or not), be reproducible, widely available, and allow measurements repeatedly over time. Ideally, biomarkers should also be non-invasive and cost-effective. This review aims to focus on the usefulness and limitations of electroencephalography (EEG) in the search for Alzheimer's disease (AD) biomarkers. The main aim of this article is to review the evolution of the most used biomarkers in AD and the need for new peripheral and, ideally, non-invasive biomarkers. The characteristics of the EEG as a possible source for biomarkers will be revised, highlighting its advantages compared to the molecular markers available so far.
Collapse
Affiliation(s)
- Paloma Monllor
- CIBERFES, Department of Physiology, Institute INCLIVA, Faculty of Medicine, Health Research University of Valencia, Avda. Blasco Ibanez 17, 46010 Valencia, Spain; (P.M.); (D.E.)
| | - Ana Cervera-Ferri
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Maria-Angeles Lloret
- Department of Clinical Neurophysiology, University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain;
| | - Daniel Esteve
- CIBERFES, Department of Physiology, Institute INCLIVA, Faculty of Medicine, Health Research University of Valencia, Avda. Blasco Ibanez 17, 46010 Valencia, Spain; (P.M.); (D.E.)
| | - Begoña Lopez
- Department of Neurology, University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain;
| | - Jose-Luis Leon
- Ascires Biomedical Group, Department of Neuroradiology, Hospital Clinico Universitario, 46010 Valencia, Spain;
| | - Ana Lloret
- CIBERFES, Department of Physiology, Institute INCLIVA, Faculty of Medicine, Health Research University of Valencia, Avda. Blasco Ibanez 17, 46010 Valencia, Spain; (P.M.); (D.E.)
| |
Collapse
|
43
|
Fu J, Zhang H, Liu S, Wu J, Zhang Y, Gao Y, Song F, Qin Y, Hu X, Liu Z. An integrated strategy using LC-MS/MS combined with in vivo microdialysis for the simultaneous determination of lignans of Schisandra chinensis (Turcz.) Baill. Fructus and endogenous neurotransmitters: application in pharmacokinetic and pharmacodynamic studies. Food Funct 2021; 12:8932-8945. [PMID: 34606559 DOI: 10.1039/d1fo01682b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Schisandra chinensis (Turcz.) Baill Fructus (SCF) is the ripe fruit of Schisandra chinensis (Turcz.) Baill, and is often used as a neuroprotective drink. Modern pharmacological studies have shown that lignans are the main bioactive components responsible for neuroprotection and have potential in the treatment of Alzheimer's disease (AD). However, the mechanism of action of SCF in the treatment of AD from the pharmacokinetics-pharmacodynamics (PK-PD) perspective remains not well established. The purpose of this study is to investigate and compare the pharmacokinetic differences of lignans in normal and AD rats, as well as to investigate their effects on neurotransmitters and their role in the treatment of AD. To achieve this goal, an integrated strategy using LC-MS/MS combined with in vivo microdialysis for the simultaneous determination of lignans of SCF and endogenous neurotransmitters has been developed and validated. The results show that the pharmacokinetic behaviors of ten lignans in the AD group were significantly different from those in the normal group. The AD group had better absorption and slower elimination than the normal group. In addition, the pharmacodynamic results of the Morris water maze (MWM) test, biochemical tests, histopathological examination, as well as immunohistochemistry analysis showed that lignans could improve the learning and memory of AD rats. The oral administration of SCF could restore the levels of the neurotransmitter parameters; seven neurotransmitters showed clockwise or counterclockwise changes with the four lignans in the hippocampal region. Taken together, the PK and PD studies based on in vivo microdialysis sampling might offer novel insights into the mechanisms of action of SCF against AD.
Collapse
Affiliation(s)
- Jun Fu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China. .,National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Hongxu Zhang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China. .,National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Shu Liu
- National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Jiajie Wu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China. .,National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Yuying Zhang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China. .,National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Yang Gao
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China. .,National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Fengrui Song
- National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Yuhua Qin
- Hainan Tropical Marine University, Sanya, 572022, China
| | - Xiuli Hu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| | - Zhongying Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
44
|
Trujillo-Estrada L, Vanderklish PW, Nguyen MMT, Kuang RR, Nguyen C, Huynh E, da Cunha C, Javonillo DI, Forner S, Martini AC, Sarraf ST, Simmon VF, Baglietto-Vargas D, LaFerla FM. SPG302 Reverses Synaptic and Cognitive Deficits Without Altering Amyloid or Tau Pathology in a Transgenic Model of Alzheimer's Disease. Neurotherapeutics 2021; 18:2468-2483. [PMID: 34738197 PMCID: PMC8804111 DOI: 10.1007/s13311-021-01143-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/04/2022] Open
Abstract
Alzheimer's disease (AD) is conceptualized as a synaptic failure disorder in which loss of glutamatergic synapses is a major driver of cognitive decline. Thus, novel therapeutic strategies aimed at regenerating synapses may represent a promising approach to mitigate cognitive deficits in AD patients. At present, no disease-modifying drugs exist for AD, and approved therapies are palliative at best, lacking in the ability to reverse the synaptic failure. Here, we tested the efficacy of a novel synaptogenic small molecule, SPG302 - a 3rd-generation benzothiazole derivative that increases the density of axospinous glutamatergic synapses - in 3xTg-AD mice. Daily dosing of 3xTg-AD mice with SPG302 at 3 and 30 mg/kg (i.p.) for 4 weeks restored hippocampal synaptic density and improved cognitive function in hippocampal-dependent tasks. Mushroom and stubby spine profiles were increased by SPG302, and associated with enhanced expression of key postsynaptic proteins - including postsynaptic density protein 95 (PSD95), drebrin, and amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) - and increased colocalization of PSD95 with synaptophysin. Notably, SPG302 proved efficacious in this model without modifying Aβ and tau pathology. Thus, our study provides preclinical support for the idea that compounds capable of restoring synaptic density offer a viable strategy to reverse cognitive decline in AD.
Collapse
Affiliation(s)
- Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- Departamento Biología Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Malaga, Spain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Peter W Vanderklish
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA.
| | - Marie Minh Thu Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Run Rong Kuang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Caroline Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Eric Huynh
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Celia da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Dominic Ibarra Javonillo
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Alessandra C Martini
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Stella T Sarraf
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA
| | - Vincent F Simmon
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA.
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, 92697-1450, USA.
- Departamento Biología Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Malaga, Spain.
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, 92697-1450, USA.
| |
Collapse
|
45
|
Singh A, Allen D, Fracassi A, Tumurbaatar B, Natarajan C, Scaduto P, Woltjer R, Kayed R, Limon A, Krishnan B, Taglialatela G. Functional Integrity of Synapses in the Central Nervous System of Cognitively Intact Individuals with High Alzheimer's Disease Neuropathology Is Associated with Absence of Synaptic Tau Oligomers. J Alzheimers Dis 2021; 78:1661-1678. [PMID: 33185603 PMCID: PMC7836055 DOI: 10.3233/jad-200716] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Certain individuals, here referred to as Non-Demented with Alzheimer Neuropathology (NDAN), do not show overt neurodegeneration (N-) and remain cognitively intact despite the presence of plaques (A+) and tangles (T+) that would normally be consistent with fully symptomatic Alzheimer's disease (AD). OBJECTIVE The existence of NDAN (A + T+N-) subjects suggests that the human brain utilizes intrinsic mechanisms that can naturally evade cognitive decline normally associated with the symptomatic stages of AD (A + T+N+). Deciphering the underlying mechanisms would prove relevant to develop complementing therapeutics to prevent progression of AD-related cognitive decline. METHODS Previously, we have reported that NDAN present with preserved neurogenesis and synaptic integrity paralleled by absence of amyloid oligomers at synapses. Using postmortem brain samples from age-matched control subjects, demented AD patients and NDAN individuals, we performed immunofluorescence, western blots, micro transplantation of synaptic membranes in Xenopus oocytes followed by twin electrode voltage clamp electrophysiology and fluorescence assisted single synaptosome-long term potentiation studies. RESULTS We report decreased tau oligomers at synapses in the brains of NDAN subjects. Furthermore, using novel approaches we report, for the first time, that such absence of tau oligomers at synapses is associated with synaptic functional integrity in NDAN subjects as compared to demented AD patients. CONCLUSION Overall, these results give further credence to tau oligomers as primary actors of synaptic destruction underscoring cognitive demise in AD and support their targeting as a viable therapeutic strategy for AD and related tauopathies.
Collapse
Affiliation(s)
- Ayush Singh
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Dyron Allen
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Batbayar Tumurbaatar
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Chandramouli Natarajan
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Pietro Scaduto
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Randy Woltjer
- Department of Pathology, Oregon Health and Science University, Portland, OR, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Agenor Limon
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA
| | - Balaji Krishnan
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA,Correspondence to: Giulio Taglialatela, PhD, Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA. Tel.: +1 409 772 1679; Fax: +1 409 772 0015; E-mail: . and Balaji Krishnan, PhD, Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA. Tel.: +1 409 772 8069; Fax: +1 409 772 0015; E-mail:
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA,Correspondence to: Giulio Taglialatela, PhD, Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA. Tel.: +1 409 772 1679; Fax: +1 409 772 0015; E-mail: . and Balaji Krishnan, PhD, Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB Galveston, TX, USA. Tel.: +1 409 772 8069; Fax: +1 409 772 0015; E-mail:
| |
Collapse
|
46
|
Tail-vein injection of MSC-derived small extracellular vesicles facilitates the restoration of hippocampal neuronal morphology and function in APP / PS1 mice. Cell Death Discov 2021; 7:230. [PMID: 34482379 PMCID: PMC8418600 DOI: 10.1038/s41420-021-00620-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/08/2021] [Accepted: 08/19/2021] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem-cell-derived small extracellular vesicles (MSC-EVs), as a therapeutic agent, have shown great promise in the treatment of neurological diseases. To date, the neurorestorative effects and underlying mechanism of MSC-EVs in Alzheimer's disease (AD) are not well known. Herein, we aimed to investigate the action of MSC-EVs on the neuronal deficits in β-amyloid protein (Aβ)-stimulated hippocampal neurons, or AD cell (SHSY5Y cell lines) and animal (APPswe / PS1dE9 mice) models. In the present study, the cell and AD models received a single-dose of MSC-EVs, and were then assessed for behavioral deficits, pathological changes, intracellular calcium transients, neuronal morphology alterations, or electrophysiological variations. Additionally, the nuclear factor E2-related factor 2 (Nrf2, a key mediator of neuronal injury in AD) signaling pathway was probed by western blotting in vitro and in vivo models of AD. Our results showed that MSC-EVs therapy improved the cognitive impairments and reduced the hippocampal Aβ aggregation and neuronal loss in AD mice. Markedly, EV treatment restored the calcium oscillations, dendritic spine alterations, action potential abnormalities, or mitochondrial changes in the hippocampus of AD models. Also, we found that the Nrf2 signaling pathway participated in the actions of MSC-EVs in the cell and animal models. Together, these data indicate that MS-EVs as promising nanotherapeutics for restoration of hippocampal neuronal morphology and function in APP / PS1 mice, further highlighting the clinical values of MSC-EVs in the treatment of AD.
Collapse
|
47
|
Yin X, Qiu Y, Zhao C, Zhou Z, Bao J, Qian W. The Role of Amyloid-Beta and Tau in the Early Pathogenesis of Alzheimer's Disease. Med Sci Monit 2021; 27:e933084. [PMID: 34471085 PMCID: PMC8422899 DOI: 10.12659/msm.933084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The abnormal accumulation of amyloid-b (Ab) and neurofibrillary tangles (NFTs) containing phosphorylated tau proteins are the main histopathological feature of Alzheimer's disease (AD). Synaptic damage and loss are earlier events than amyloid plaques and NFTs in AD progress and best correlate with cognitive deficits in AD patients. Soluble oligomeric Aß initiates the progression of AD and tau mediates the subsequent synaptic impairments at an early stage of AD. In this review we discuss how Ab or/and tau causes synaptic dysfunction. Ab oligomers gather at synapses and give rise to synaptic death in a variety of ways such as regulating receptors and receptor tyrosine kinases, unbalancing calcium homeostasis, and activating caspases and calcineurin. A large amount of hyperphosphorylated tau exists in the synapse of the AD brain. Aß-triggered synaptic deficits are dependent on tau. Soluble, hyperphosphorylated tau is much more correlated to cognitive decline in AD patients. Tau-targeted therapies have received more attention because the treatments targeting Aß failed in AD. Here, we also review the therapy strategies used to intervene in the very early stages of AD. Soluble hyperphosphorylated tau forms a complex with cell surface receptors, scaffold proteins, or intracellular signaling molecules to damage synaptic function. Therefore, therapeutic strategies targeting synaptic tau at the early stage of AD may ameliorating pathology in AD. This review aims to provide an update on the role of oligomeric Ab and soluble hyperphosphorylated tau in the early pathogenesis of Alzheimer's disease and to develop a new treatment strategy based on this.
Collapse
Affiliation(s)
- Xiaomin Yin
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland).,Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China (mainland).,NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Yanyan Qiu
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Chenhao Zhao
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Zheng Zhou
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Junze Bao
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Wei Qian
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland).,Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China (mainland).,NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China (mainland)
| |
Collapse
|
48
|
Bréchet L, Michel CM, Schacter DL, Pascual-Leone A. Improving autobiographical memory in Alzheimer's disease by transcranial alternating current stimulation. Curr Opin Behav Sci 2021; 40:64-71. [PMID: 34485630 PMCID: PMC8415489 DOI: 10.1016/j.cobeha.2021.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We review the latest evidence from animal models, studies in humans using electrophysiology, experimental memory paradigms, and non-invasive brain stimulation (NIBS), in the form of transcranial alternating current stimulation (tACS), suggesting that the altered activity in networks that contribute to the autobiographical memory (ABM) deficits may be modifiable. ABM involves a specific brain network of interacting regions that store and retrieve life experiences. Deficits in ABM are early symptoms in patients with Alzheimer's disease (AD), and serve as relevant predictors of disease progression. The possibility to modify the neural substrates of ABM opens exciting avenues for the development of therapeutic approaches. Beyond a summary of the causal role of brain oscillations in ABM, we propose a new approach of modulating brain oscillations using personalized tACS with the possibility of reducing ABM deficits. We suggest that human experimental studies using cognitive tasks, EEG, and tACS can have future translational clinical implications.
Collapse
Affiliation(s)
- Lucie Bréchet
- Hinda and Arthur Marcus Institute for Aging Research and Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, Massachusetts, USA
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
- Center for Biomedical Imaging (CIBM), Lausanne, Geneva, Switzerland
| | - Christoph M. Michel
- Functional Brain Mapping Laboratory, Fundamental Neuroscience Dept., University Geneva, Switzerland
- Center for Biomedical Imaging (CIBM), Lausanne, Geneva, Switzerland
| | | | - Alvaro Pascual-Leone
- Hinda and Arthur Marcus Institute for Aging Research and Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, Massachusetts, USA
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
- Guttmann Brain Health Institute, Institut Guttman de Neurorehabilitació, Barcelona, Spain
| |
Collapse
|
49
|
Xu D, Yang P, Yang ZJ, Li QG, Ouyang YT, Yu T, Shangguan JH, Wan YY, Jiang LP, Qu XH, Han XJ. Blockage of Drp1 phosphorylation at Ser579 protects neurons against Aβ 1‑42‑induced degeneration. Mol Med Rep 2021; 24:657. [PMID: 34278489 PMCID: PMC8299198 DOI: 10.3892/mmr.2021.12296] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/24/2021] [Indexed: 01/13/2023] Open
Abstract
Alzheimer's disease (AD), one of the most common types of chronic neurodegenerative diseases, is pathologically characterized by the formation of amyloid β (Aβ) peptide-containing plaques and neurofibrillary tangles. Among Aβ peptides, Aβ1–42 induces neuronal toxicity and neurodegeneration. In our previous studies, Cdk5 was found to regulate Aβ1–42-induced mitochondrial fission via the phosphorylation of dynamin-related protein 1 (Drp1) at Ser579. However, whether blockage of Drp1 phosphorylation at Ser579 protects neurons against Aβ1–42-induced degeneration remains to be elucidated. Thus, the aim the present study was to examine the effect of mutant Drp1-S579A on neurodegeneration and its underlying mechanism. First, the phosphorylation-defect (phospho-defect) mutant, Lenti-Drp1-S579A was constructed. Phospho-defect Drp1-S579A expression was detected in primary cultures of mouse cortical neurons infected with Lenti-Drp1-S579A using western blotting and it was found to successfully attenuate the phosphorylation of endogenous Drp1 at Ser579. In primary neuronal cultures, the neuronal processes were evaluated under microscopy. Treatment with 10 µM Aβ1–42 significantly decreased dendritic density and length, spine outgrowth and synapse number. As expected, infection of neurons with Lenti-Drp1-S579A efficiently alleviated the inhibitory effect of Aβ1–42 on neurite outgrowth and synapse density. In addition, infection with Lenti-Drp1-S579A abolished the cleavage of caspase-3 and apoptosis in neurons exposed to Aβ1–42. Thus, the current data demonstrated that blockage of Drp1 phosphorylation at Ser579 may be an effective strategy to protect neurons against Aβ1–42-induced degeneration and apoptosis. These findings underline the therapeutic potential of targeting Drp1 in the treatment of AD.
Collapse
Affiliation(s)
- Dan Xu
- Institute of Geriatrics, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ping Yang
- Department of Neurology, Affiliated People's Hospital of Nanchang University, Jiangxi 330006, P.R. China
| | - Zhang-Jian Yang
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Jiangxi 330006, P.R. China
| | - Qiu-Gen Li
- Institute of Geriatrics, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ye-Tong Ouyang
- Department of Neurology, Affiliated People's Hospital of Nanchang University, Jiangxi 330006, P.R. China
| | - Ting Yu
- Department of Neurology, Affiliated People's Hospital of Nanchang University, Jiangxi 330006, P.R. China
| | - Jian-Hui Shangguan
- Department of Neurology, Affiliated People's Hospital of Nanchang University, Jiangxi 330006, P.R. China
| | - Yu-Ying Wan
- Department of Intra‑Hospital Infection Management, The Second Affiliated Hospital of Nanchang University, Jiangxi 330006, P.R. China
| | - Li-Ping Jiang
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Jiangxi 330006, P.R. China
| | - Xin-Hui Qu
- Institute of Geriatrics, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao-Jian Han
- Institute of Geriatrics, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
50
|
Gasiorowska A, Wydrych M, Drapich P, Zadrozny M, Steczkowska M, Niewiadomski W, Niewiadomska G. The Biology and Pathobiology of Glutamatergic, Cholinergic, and Dopaminergic Signaling in the Aging Brain. Front Aging Neurosci 2021; 13:654931. [PMID: 34326765 PMCID: PMC8315271 DOI: 10.3389/fnagi.2021.654931] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
The elderly population is growing worldwide, with important health and socioeconomic implications. Clinical and experimental studies on aging have uncovered numerous changes in the brain, such as decreased neurogenesis, increased synaptic defects, greater metabolic stress, and enhanced inflammation. These changes are associated with cognitive decline and neurobehavioral deficits. Although aging is not a disease, it is a significant risk factor for functional worsening, affective impairment, disease exaggeration, dementia, and general disease susceptibility. Conversely, life events related to mental stress and trauma can also lead to accelerated age-associated disorders and dementia. Here, we review human studies and studies on mice and rats, such as those modeling human neurodegenerative diseases, that have helped elucidate (1) the dynamics and mechanisms underlying the biological and pathological aging of the main projecting systems in the brain (glutamatergic, cholinergic, and dopaminergic) and (2) the effect of defective glutamatergic, cholinergic, and dopaminergic projection on disabilities associated with aging and neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. Detailed knowledge of the mechanisms of age-related diseases can be an important element in the development of effective ways of treatment. In this context, we briefly analyze which adverse changes associated with neurodegenerative diseases in the cholinergic, glutaminergic and dopaminergic systems could be targeted by therapeutic strategies developed as a result of our better understanding of these damaging mechanisms.
Collapse
Affiliation(s)
- Anna Gasiorowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Wydrych
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Patrycja Drapich
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Maciej Zadrozny
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Marta Steczkowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Wiktor Niewiadomski
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Grazyna Niewiadomska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|