1
|
Higa GSV, Viana FJC, Francis-Oliveira J, Cruvinel E, Franchin TS, Marcourakis T, Ulrich H, De Pasquale R. Serotonergic neuromodulation of synaptic plasticity. Neuropharmacology 2024; 257:110036. [PMID: 38876308 DOI: 10.1016/j.neuropharm.2024.110036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Synaptic plasticity constitutes a fundamental process in the reorganization of neural networks that underlie memory, cognition, emotional responses, and behavioral planning. At the core of this phenomenon lie Hebbian mechanisms, wherein frequent synaptic stimulation induces long-term potentiation (LTP), while less activation leads to long-term depression (LTD). The synaptic reorganization of neuronal networks is regulated by serotonin (5-HT), a neuromodulator capable of modify synaptic plasticity to appropriately respond to mental and behavioral states, such as alertness, attention, concentration, motivation, and mood. Lately, understanding the serotonergic Neuromodulation of synaptic plasticity has become imperative for unraveling its impact on cognitive, emotional, and behavioral functions. Through a comparative analysis across three main forebrain structures-the hippocampus, amygdala, and prefrontal cortex, this review discusses the actions of 5-HT on synaptic plasticity, offering insights into its role as a neuromodulator involved in emotional and cognitive functions. By distinguishing between plastic and metaplastic effects, we provide a comprehensive overview about the mechanisms of 5-HT neuromodulation of synaptic plasticity and associated functions across different brain regions.
Collapse
Affiliation(s)
- Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - José Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Thainá Soares Franchin
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Tania Marcourakis
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
2
|
Hu NW, Ondrejcak T, Klyubin I, Yang Y, Walsh DM, Livesey FJ, Rowan MJ. Patient-derived tau and amyloid-β facilitate long-term depression in vivo: role of tumour necrosis factor-α and the integrated stress response. Brain Commun 2024; 6:fcae333. [PMID: 39391333 PMCID: PMC11465085 DOI: 10.1093/braincomms/fcae333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 08/22/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease is characterized by a progressive cognitive decline in older individuals accompanied by the deposition of two pathognomonic proteins amyloid-β and tau. It is well documented that synaptotoxic soluble amyloid-β aggregates facilitate synaptic long-term depression, a major form of synaptic weakening that correlates with cognitive status in Alzheimer's disease. Whether synaptotoxic tau, which is also associated strongly with progressive cognitive decline in patients with Alzheimer's disease and other tauopathies, also causes facilitation remains to be clarified. Young male adult and middle-aged rats were employed. Synaptotoxic tau and amyloid-β were obtained from different sources including (i) aqueous brain extracts from patients with Alzheimer's disease and Pick's disease tauopathy; (ii) the secretomes of induced pluripotent stem cell-derived neurons from individuals with trisomy of chromosome 21; and (iii) synthetic amyloid-β. In vivo electrophysiology was performed in urethane anaesthetized animals. Evoked field excitatory postsynaptic potentials were recorded from the stratum radiatum in the CA1 area of the hippocampus with electrical stimulation to the Schaffer collateral-commissural pathway. To study the enhancement of long-term depression, relatively weak low-frequency electrical stimulation was used to trigger peri-threshold long-term depression. Synaptotoxic forms of tau or amyloid-β were administered intracerebroventricularly. The ability of agents that inhibit the cytokine tumour necrosis factor-α or the integrated stress response to prevent the effects of amyloid-β or tau on long-term depression was assessed after local or systemic injection, respectively. We found that diffusible tau from Alzheimer's disease or Pick's disease patients' brain aqueous extracts or the secretomes of trisomy of chromosome 21 induced pluripotent stem cell-derived neurons, like Alzheimer's disease brain-derived amyloid-β and synthetic oligomeric amyloid-β, potently enhanced synaptic long-term depression in live rats. We further demonstrated that long-term depression facilitation by both tau and amyloid-β was age-dependent, being more potent in middle-aged compared with young animals. Finally, at the cellular level, we provide pharmacological evidence that tumour necrosis factor-α and the integrated stress response are downstream mediators of long-term depression facilitation by both synaptotoxic tau and amyloid-β. Overall, these findings reveal the promotion of an age-dependent synaptic weakening by both synaptotoxic tau and amyloid-β. Pharmacologically targeting shared mechanisms of tau and amyloid-β synaptotoxicity, such as tumour necrosis factor-α or the integrated stress response, provides an attractive strategy to treat early Alzheimer's disease.
Collapse
Affiliation(s)
- Neng-Wei Hu
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Dublin, Ireland
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Tomas Ondrejcak
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Dublin, Ireland
| | - Igor Klyubin
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Dublin, Ireland
| | - Yin Yang
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Dublin, Ireland
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Frederick J Livesey
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, London WC1N 1DZ, UK
| | - Michael J Rowan
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Dublin, Ireland
| |
Collapse
|
3
|
Sánchez-Fernández N, Gómez-Acero L, Castañé A, Adell A, Campa L, Bonaventura J, Brito V, Ginés S, Queiróz F, Silva H, Lopes JP, Lopes CR, Radošević M, Gasull X, Cunha RA, Köfalvi A, Ferreira SG, Ciruela F, Aso E. A combination of Δ 9-tetrahydrocannabinol and cannabidiol modulates glutamate dynamics in the hippocampus of an animal model of Alzheimer's disease. Neurotherapeutics 2024; 21:e00439. [PMID: 39232876 DOI: 10.1016/j.neurot.2024.e00439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024] Open
Abstract
A combination of Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD) at non-psychoactive doses was previously demonstrated to reduce cognitive decline in APP/PS1 mice, an animal model of Alzheimer's disease (AD). However, the neurobiological substrates underlying these therapeutic properties of Δ9-THC and CBD are not fully understood. Considering that dysregulation of glutamatergic activity contributes to cognitive impairment in AD, the present study evaluates the hypothesis that the combination of these two natural cannabinoids might reverse the alterations in glutamate dynamics within the hippocampus of this animal model of AD. Interestingly, our findings reveal that chronic treatment with Δ9-THC and CBD, but not with any of them alone, reduces extracellular glutamate levels and the basal excitability of the hippocampus in APP/PS1 mice. These effects are not related to significant changes in the function and structure of glutamate synapses, as no relevant changes in synaptic plasticity, glutamate signaling or in the levels of key components of these synapses were observed in cannabinoid-treated mice. Our data instead indicate that these cannabinoid effects are associated with the control of glutamate uptake and/or to the regulation of the hippocampal network. Taken together, these results support the potential therapeutic properties of combining these natural cannabinoids against the excitotoxicity that occurs in AD brains.
Collapse
Affiliation(s)
- Nuria Sánchez-Fernández
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L'Hospitalet de Llobregat, Spain
| | - Laura Gómez-Acero
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L'Hospitalet de Llobregat, Spain
| | - Anna Castañé
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), 08036 Barcelona, Spain; Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain; Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), 08500 Vic, Spain
| | - Albert Adell
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain; Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Cantabria, 39011 Santander, Spain
| | - Leticia Campa
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Jordi Bonaventura
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L'Hospitalet de Llobregat, Spain
| | - Verónica Brito
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - Silvia Ginés
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - Francisco Queiróz
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Henrique Silva
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - João Pedro Lopes
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Cátia R Lopes
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Marija Radošević
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Xavier Gasull
- Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; FMUC-Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Attila Köfalvi
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Samira G Ferreira
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L'Hospitalet de Llobregat, Spain
| | - Ester Aso
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
4
|
Lu MN, Wang D, Ye CJ, Yan GJ, Song JF, Shi XY, Li SS, Liu LN, Zhang HX, Dong XH, Hu T, Wang XY, Xiyang YB. Navβ2 Intracellular Fragments Contribute to Aβ1-42-Induced Cognitive Impairment and Synaptic Deficit Through Transcriptional Suppression of BDNF. Mol Neurobiol 2024:10.1007/s12035-024-04317-y. [PMID: 38965172 DOI: 10.1007/s12035-024-04317-y] [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: 02/17/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
A pathological hallmark of Alzheimer's disease (AD) is the region-specific accumulation of the amyloid-beta protein (Aβ), which triggers aberrant neuronal excitability, synaptic impairment, and progressive cognitive decline. Previous works have demonstrated that Aβ pathology induced aberrant elevation in the levels and excessive enzymatic hydrolysis of voltage-gated sodium channel type 2 beta subunit (Navβ2) in the brain of AD models, accompanied by alteration in excitability of hippocampal neurons, synaptic deficits, and subsequently, cognitive dysfunction. However, the mechanism is unclear. In this research, by employing cell models treated with toxic Aβ1-42 and AD mice, the possible effects and potential mechanisms induced by Navβ2. The results reveal that Aβ1-42 induces remarkable increases in Navβ2 intracellular domain (Navβ2-ICD) and decreases in both BDNF exons and protein levels, as well as phosphorylated tropomyosin-related kinase B (pTrkB) expression in cells and mice, coupled with cognitive impairments, synaptic deficits, and aberrant neuronal excitability. Administration with exogenous Navβ2-ICD further enhances these effects induced by Aβ1-42, while interfering the generation of Navβ2-ICD and/or complementing BDNF neutralize the Navβ2-ICD-conducted effects. Luciferase reporter assay verifies that Navβ2-ICD regulates BDNF transcription and expression by targeting its promoter. Collectively, our findings partially elucidate that abnormal enzymatic hydrolysis of Navβ2 induced by Aβ1-42-associated AD pathology leads to intracellular Navβ2-ICD overload, which may responsible to abnormal neuronal excitability, synaptic deficit, and cognition dysfunction, through its transcriptional suppression on BDNF. Therefore, this work supplies novel evidences that Navβ2 plays crucial roles in the occurrence and progression of cognitive impairment of AD by transcriptional regulatory activity of its cleaved ICD.
Collapse
Affiliation(s)
- Min-Nan Lu
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Dan Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Chen-Jun Ye
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Guo-Ji Yan
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jing-Feng Song
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xin-Ying Shi
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Shan-Shan Li
- Experimental Teaching Center, Basic Medical College, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Li-Na Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Hui-Xiang Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xiao-Han Dong
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Tao Hu
- Department of Laboratory Medicine, The Third People's Hospital of Yunnan Province, Kunming, 650000, Yunnan, China
| | - Xu-Yang Wang
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yan-Bin Xiyang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
5
|
Hafycz JM, Strus E, Naidoo NN. Early and late chaperone intervention therapy boosts XBP1s and ADAM10, restores proteostasis, and rescues learning in Alzheimer's Disease mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541973. [PMID: 37292838 PMCID: PMC10245863 DOI: 10.1101/2023.05.23.541973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder that is pervasive among the aging population. Two distinct phenotypes of AD are deficits in cognition and proteostasis, including chronic activation of the unfolded protein response (UPR) and aberrant Aβ production. It is unknown if restoring proteostasis by reducing chronic and aberrant UPR activation in AD can improve pathology and cognition. Here, we present data using an APP knock-in mouse model of AD and several protein chaperone supplementation paradigms, including a late-stage intervention. We show that supplementing protein chaperones systemically and locally in the hippocampus reduces PERK signaling and increases XBP1s, which is associated with increased ADAM10 and decreased Aβ42. Importantly, chaperone treatment improves cognition which is correlated with increased CREB phosphorylation and BDNF. Together, this data suggests that chaperone treatment restores proteostasis in a mouse model of AD and that this restoration is associated with improved cognition and reduced pathology. One-sentence summary Chaperone therapy in a mouse model of Alzheimer's disease improves cognition by reducing chronic UPR activity.
Collapse
|
6
|
Valdivia G, Ardiles AO, Idowu A, Salazar C, Lee HK, Gallagher M, Palacios AG, Kirkwood A. mGluR-dependent plasticity in rodent models of Alzheimer's disease. Front Synaptic Neurosci 2023; 15:1123294. [PMID: 36937569 PMCID: PMC10017879 DOI: 10.3389/fnsyn.2023.1123294] [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: 12/13/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Long-term potentiation (LTP) and depression (LTD) are currently the most comprehensive models of synaptic plasticity models to subserve learning and memory. In the CA1 region of the hippocampus LTP and LTD can be induced by the activation of either NMDA receptors or mGluR5 metabotropic glutamate receptors. Alterations in either form of synaptic plasticity, NMDAR-dependent or mGluR-dependent, are attractive candidates to contribute to learning deficits in conditions like Alzheimer's disease (AD) and aging. Research, however, has focused predominantly on NMDAR-dependent forms of LTP and LTD. Here we studied age-associated changes in mGluR-dependent LTP and LTD in the APP/PS1 mouse model of AD and in Octodon degu, a rodent model of aging that exhibits features of AD. At 2 months of age, APP/PS1 mouse exhibited robust mGluR-dependent LTP and LTD that was completely lost by the 8th month of age. The expression of mGluR protein in the hippocampus of APP/PS1 mice was not affected, consistent with previous findings indicating the uncoupling of the plasticity cascade from mGluR5 activation. In O. degu, the average mGluR-LTD magnitude is reduced by half by the 3 rd year of age. In aged O. degu individuals, the reduced mGluR-LTD correlated with reduced performance in a radial arm maze task. Altogether these findings support the idea that the preservation of mGluR-dependent synaptic plasticity is essential for the preservation of learning capacity during aging.
Collapse
Affiliation(s)
- Gonzalo Valdivia
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, Baltimore, MD, United States
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Alvaro O. Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Abimbola Idowu
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, Baltimore, MD, United States
| | - Claudia Salazar
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Hey-Kyoung Lee
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, Baltimore, MD, United States
| | - Michela Gallagher
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Adrian G. Palacios
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Alfredo Kirkwood
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
7
|
Bioactive human Alzheimer brain soluble Aβ: pathophysiology and therapeutic opportunities. Mol Psychiatry 2022; 27:3182-3191. [PMID: 35484241 DOI: 10.1038/s41380-022-01589-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 12/16/2022]
Abstract
The accumulation of amyloid-β protein (Aβ) plays an early role in the pathogenesis of Alzheimer's disease (AD). The precise mechanism of how Aβ accumulation leads to synaptic dysfunction and cognitive impairment remains unclear but is likely due to small soluble oligomers of Aβ (oAβ). Most studies have used chemical synthetic or cell-secreted Aβ oligomers to study their pathogenic mechanisms, but the Aβ derived from human AD brain tissue is less well characterized. Here we review updated knowledge on the extraction and characterization of bioactive human AD brain oAβ and the mechanisms by which they cause hippocampal synaptic dysfunction. Human AD brain-derived oAβ can impair hippocampal long-term potentiation (LTP) and enhance long-term depression (LTD). Many studies suggest that oAβ may directly disrupt neuronal NMDA receptors, AMPA receptors and metabotropic glutamate receptors (mGluRs). oAβ also impairs astrocytic synaptic functions, including glutamate uptake, D-serine release, and NMDA receptor function. We also discuss oAβ-induced neuronal hyperexcitation. These results may suggest a multi-target approach for the treatment of AD, including both oAβ neutralization and reversal of glutamate-mediated excitotoxicity.
Collapse
|
8
|
Krafft GA, Jerecic J, Siemers E, Cline EN. ACU193: An Immunotherapeutic Poised to Test the Amyloid β Oligomer Hypothesis of Alzheimer’s Disease. Front Neurosci 2022; 16:848215. [PMID: 35557606 PMCID: PMC9088393 DOI: 10.3389/fnins.2022.848215] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/02/2022] [Indexed: 12/26/2022] Open
Abstract
Alzheimer’s disease (AD) is an age-related neurodegenerative disease that affects 50 million people worldwide, with 10 million new cases occurring each year. The emotional and economic impacts of AD on patients and families are devastating. Approved treatments confer modest improvement in symptoms, and recently one treatment obtained accelerated approval from the United States Food and Drug Administration (FDA) and may have modest disease modifying benefit. Research over the past three decades has established a clear causal linkage between AD and elevated brain levels of amyloid β (Aβ) peptide, and substantial evidence now implicates soluble, non-fibrillar Aβ oligomers (AβOs) as the molecular assemblies directly responsible for AD-associated memory and cognitive failure and accompanying progressive neurodegeneration. The widely recognized linkage of elevated Aβ and AD spawned a comprehensive 20-year therapeutic campaign that focused primarily on two strategies – inhibition of the secretase enzymes responsible for Aβ production and clearance of Aβ peptide or amyloid plaques with Aβ-directed immunotherapeutics. Unfortunately, all clinical trials of secretase inhibitors were unsuccessful. Of the completed phase 3 immunotherapy programs, bapineuzumab (targeting amyloid plaque) and solanezumab (targeting Aβ monomers) were negative, and the crenezumab program (targeting Aβ monomers and to a small extent oligomers) was stopped for futility. Aducanumab (targeting amyloid plaques), which recently received FDA accelerated approval, had one positive and one negative phase 3 trial. More than 25 negative randomized clinical trials (RCTs) have evaluated Aβ-targeting therapeutics, yet none has directly evaluated whether selective blockage of disease-relevant AβOs can stop or reverse AD-associated cognitive decline. Here, we briefly summarize studies that establish the AD therapeutic rationale to target AβOs selectively, and we describe ACU193, the first AβO-selective immunotherapeutic to enter human clinical trials and the first positioned to test the AβO hypothesis of AD.
Collapse
|
9
|
Shobo A, Röntgen A, Hancock MA, Multhaup G. Biophysical characterization as a tool to predict amyloidogenic and toxic properties of amyloid-β42 peptides. FEBS Lett 2022; 596:1401-1411. [PMID: 35466397 DOI: 10.1002/1873-3468.14358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/02/2022] [Accepted: 04/19/2022] [Indexed: 11/06/2022]
Abstract
Amyloid-β42 (Aβ42) peptides are central to the amyloid pathology in Alzheimer's disease (AD). As biological mimetics, properties of synthetic Aβ peptides usually vary between vendors and batches, thus impacting the reproducibility of experimental studies. Here, we tested recombinantly expressed Aβ42 (Asp1 to Ala42) against synthetic Aβ42 from different suppliers using matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS), circular dichroism (CD) spectroscopy, thioflavin T aggregation, surface plasmon resonance and MTT cell viability assays. Overall, our recombinant Aβ42 provided a reproducible mimetic of desired properties. Across experimental approaches, the combined detection of Aβ42 dimers and random coil to β-sheet transition only correlated with aggregation-prone and cytotoxic peptides. Conclusively, combining MALDI-MS with CD appears to provide a rapid, reliable means to predict the "bioactivity" of Aβ42.
Collapse
Affiliation(s)
- Adeola Shobo
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | - Alexander Röntgen
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada.,Institute of Biochemistry, University of Cologne, Cologne, Germany
| | - Mark A Hancock
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada.,SPR-MS Facility, McGill University, Montreal, QC, Canada
| | - Gerhard Multhaup
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| |
Collapse
|
10
|
Modulation of Amyloid β-Induced Microglia Activation and Neuronal Cell Death by Curcumin and Analogues. Int J Mol Sci 2022; 23:ijms23084381. [PMID: 35457197 PMCID: PMC9027876 DOI: 10.3390/ijms23084381] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that is not restricted to the neuronal compartment but includes important interactions with immune cells, including microglia. Protein aggregates, common pathological hallmarks of AD, bind to pattern recognition receptors on microglia and trigger an inflammatory response, which contributes to disease progression and severity. In this context, curcumin is emerging as a potential drug candidate able to affect multiple key pathways implicated in AD, including neuroinflammation. Therefore, we studied the effect of curcumin and its structurally related analogues cur6 and cur16 on amyloid-β (Aβ)-induced microglia activation and neuronal cell death, as well as their effect on the modulation of Aβ aggregation. Primary cortical microglia and neurons were exposed to two different populations of Aβ42 oligomers (Aβ42Os) where the oligomeric state had been assigned by capillary electrophoresis and ultrafiltration. When stimulated with high molecular weight Aβ42Os, microglia released proinflammatory cytokines that led to early neuronal cell death. The studied compounds exerted an anti-inflammatory effect on high molecular weight Aβ42O-stimulated microglia and possibly inhibited microglia-mediated neuronal cell toxicity. Furthermore, the tested compounds demonstrated antioligomeric activity during the process of in vitro Aβ42 aggregation. These findings could be investigated further and used for the optimization of multipotent candidate molecules for AD treatment.
Collapse
|
11
|
Shipton OA, Tang CS, Paulsen O, Vargas-Caballero M. Differential vulnerability of hippocampal CA3-CA1 synapses to Aβ. Acta Neuropathol Commun 2022; 10:45. [PMID: 35379353 PMCID: PMC8981624 DOI: 10.1186/s40478-022-01350-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/18/2022] [Indexed: 11/25/2022] Open
Abstract
Amyloid-beta (Aβ) and tau protein are both involved in the pathogenesis of Alzheimer’s disease. Aβ produces synaptic deficits in wild-type mice that are not seen in Mapt−/− mice, suggesting that tau protein is required for these effects of Aβ. However, whether some synapses are more selectively affected and what factors may determine synaptic vulnerability to Aβ are poorly understood. Here we first observed that burst timing-dependent long-term potentiation (b-LTP) in hippocampal CA3-CA1 synapses, which requires GluN2B subunit-containing NMDA receptors (NMDARs), was inhibited by human Aβ1–42 (hAβ) in wild-type (WT) mice, but not in tau-knockout (Mapt−/−) mice. We then tested whether NMDAR currents were affected by hAβ; we found that hAβ reduced the postsynaptic NMDAR current in WT mice but not in Mapt−/− mice, while the NMDAR current was reduced to a similar extent by the GluN2B-selective NMDAR antagonist Ro 25–6981. To further investigate a possible difference in GluN2B-containing NMDARs in Mapt−/− mice, we used optogenetics to compare NMDAR/AMPAR ratio of EPSCs in CA1 synapses with input from left vs right CA3. It was previously reported in WT mice that hippocampal synapses in CA1 that receive input from the left CA3 display a higher NMDAR charge transfer and a higher Ro-sensitivity than synapses in CA1 that receive input from the right CA3. Here we observed the same pattern in Mapt−/− mice, thus differential NMDAR subunit expression does not explain the difference in hAβ effect on LTP. Finally, we asked whether synapses with left vs right CA3 input are differentially affected by hAβ in WT mice. We found that NMDAR current in synapses with input from the left CA3 were reduced while synapses with input from the right CA3 were unaffected by acute hAβ exposure. These results suggest that hippocampal CA3-CA1 synapses with presynaptic axon originating in the left CA3 are selectively vulnerable to Aβ and that a genetic knock out of tau protein protects them from Aβ synaptotoxicity.
Collapse
|
12
|
Electrochemical aptamer-based nanobiosensors for diagnosing Alzheimer's disease: A review. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 135:112689. [DOI: 10.1016/j.msec.2022.112689] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 12/22/2022]
|
13
|
Liu Y, Ding R, Xu Z, Xue Y, Zhang D, Zhang Y, Li W, Li X. Roles and Mechanisms of the Protein Quality Control System in Alzheimer's Disease. Int J Mol Sci 2021; 23:345. [PMID: 35008771 PMCID: PMC8745298 DOI: 10.3390/ijms23010345] [Citation(s) in RCA: 4] [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] [Received: 11/30/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the deposition of senile plaques (SPs) and the formation of neurofibrillary tangles (NTFs), as well as neuronal dysfunctions in the brain, but in fact, patients have shown a sustained disease progression for at least 10 to 15 years before these pathologic biomarkers can be detected. Consequently, as the most common chronic neurological disease in the elderly, the challenge of AD treatment is that it is short of effective biomarkers for early diagnosis. The protein quality control system is a collection of cellular pathways that can recognize damaged proteins and thereby modulate their turnover. Abundant evidence indicates that the accumulation of abnormal proteins in AD is closely related to the dysfunction of the protein quality control system. In particular, it is the synthesis, degradation, and removal of essential biological components that have already changed in the early stage of AD, which further encourages us to pay more attention to the protein quality control system. The review mainly focuses on the endoplasmic reticulum system (ERS), autophagy-lysosome system (ALS) and the ubiquitin-proteasome system (UPS), and deeply discusses the relationship between the protein quality control system and the abnormal proteins of AD, which can not only help us to understand how and why the complex regulatory system becomes malfunctional during AD progression, but also provide more novel therapeutic strategies to prevent the development of AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xing Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (Y.L.); (R.D.); (Z.X.); (Y.X.); (D.Z.); (Y.Z.); (W.L.)
| |
Collapse
|
14
|
The Density of Group I mGlu 5 Receptors Is Reduced along the Neuronal Surface of Hippocampal Cells in a Mouse Model of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22115867. [PMID: 34070808 PMCID: PMC8199018 DOI: 10.3390/ijms22115867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 11/17/2022] Open
Abstract
Metabotropic glutamate receptor subtype 5 (mGlu5) is implicated in the pathophysiology of Alzheimer’s disease (AD). However, its alteration at the subcellular level in neurons is still unexplored. Here, we provide a quantitative description on the expression and localisation patterns of mGlu5 in the APP/PS1 model of AD at 12 months of age, combining immunoblots, histoblots and high-resolution immunoelectron microscopic approaches. Immunoblots revealed that the total amount of mGlu5 protein in the hippocampus, in addition to downstream molecules, i.e., Gq/11 and PLCβ1, was similar in both APP/PS1 mice and age-matched wild type mice. Histoblots revealed that mGlu5 expression in the brain and its laminar expression in the hippocampus was also unaltered. However, the ultrastructural techniques of SDS-FRL and pre-embedding immunogold demonstrated that the subcellular localisation of mGlu5 was significantly reduced along the neuronal surface of hippocampal principal cells, including CA1 pyramidal cells and DG granule cells, in APP/PS1 mice at 12 months of age. The decrease in the surface localisation of mGlu5 was accompanied by an increase in its frequency at intracellular sites in the two neuronal populations. Together, these data demonstrate, for the first time, a loss of mGlu5 at the plasma membrane and accumulation at intracellular sites in different principal cells of the hippocampus in APP/PS1 mice, suggesting an alteration of the excitability and synaptic transmission that could contribute to the cognitive dysfunctions in this AD animal model. Further studies are required to elucidate the specificity of mGlu5-associated molecules and downstream signalling pathways in the progression of the pathology.
Collapse
|
15
|
Araya-Arriagada J, Bello F, Shivashankar G, Neira D, Durán-Aniotz C, Acosta ML, Escobar MJ, Hetz C, Chacón M, Palacios AG. Retinal Ganglion Cells Functional Changes in a Mouse Model of Alzheimer's Disease Are Linked with Neurotransmitter Alterations. J Alzheimers Dis 2021; 82:S5-S18. [PMID: 33749647 DOI: 10.3233/jad-201195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent form of dementia worldwide. This neurodegenerative syndrome affects cognition, memory, behavior, and the visual system, particularly the retina. OBJECTIVE This work aims to determine whether the 5xFAD mouse, a transgenic model of AD, displays changes in the function of retinal ganglion cells (RGCs) and if those alterations are correlated with changes in the expression of glutamate and gamma-aminobutyric acid (GABA) neurotransmitters. METHODS In young (2-3-month-old) and adult (6-7-month-old) 5xFAD and WT mice, we have studied the physiological response, firing rate, and burst of RGCs to various types of visual stimuli using a multielectrode array system. RESULTS The firing rate and burst response in 5xFAD RGCs showed hyperactivity at the early stage of AD in young mice, whereas hypoactivity was seen at the later stage of AD in adults. The physiological alterations observed in 5xFAD correlate well with an increase in the expression of glutamate in the ganglion cell layer in young and adults. GABA staining increased in the inner nuclear and plexiform layer, which was more pronounced in the adult than the young 5xFAD retina, altering the excitation/inhibition balance, which could explain the observed early hyperactivity and later hypoactivity in RGC physiology. CONCLUSION These findings indicate functional changes may be caused by neurochemical alterations of the retina starting at an early stage of the AD disease.
Collapse
Affiliation(s)
- Joaquín Araya-Arriagada
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Chile
| | - Felipe Bello
- Department of Engineering Informatics, Universidad de Santiago, Santiago, Chile
| | - Gaganashree Shivashankar
- School of Optometry and Vision Science; Centre for Brain Research; Brain Research New Zealand; The University of Auckland, Auckland, New Zealand
| | - David Neira
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudia Durán-Aniotz
- Center for Social and Cognitive Neuroscience, School of Psychology, Universidad Adolfo Ibáñez, Santiago de Chile, Chile
| | - Mónica L Acosta
- School of Optometry and Vision Science; Centre for Brain Research; Brain Research New Zealand; The University of Auckland, Auckland, New Zealand
| | - María José Escobar
- Departamento de Electrónica, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile
| | - Max Chacón
- Department of Engineering Informatics, Universidad de Santiago, Santiago, Chile
| | - Adrián G Palacios
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
16
|
Nagy D, Ennis KA, Wei R, Su SC, Hinckley CA, Gu RF, Gao B, Massol RH, Ehrenfels C, Jandreski L, Thomas AM, Nelson A, Gyoneva S, Hajós M, Burkly LC. Developmental synaptic regulator, TWEAK/Fn14 signaling, is a determinant of synaptic function in models of stroke and neurodegeneration. Proc Natl Acad Sci U S A 2021; 118:e2001679118. [PMID: 33526652 PMCID: PMC8017933 DOI: 10.1073/pnas.2001679118] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Identifying molecular mediators of neural circuit development and/or function that contribute to circuit dysfunction when aberrantly reengaged in neurological disorders is of high importance. The role of the TWEAK/Fn14 pathway, which was recently reported to be a microglial/neuronal axis mediating synaptic refinement in experience-dependent visual development, has not been explored in synaptic function within the mature central nervous system. By combining electrophysiological and phosphoproteomic approaches, we show that TWEAK acutely dampens basal synaptic transmission and plasticity through neuronal Fn14 and impacts the phosphorylation state of pre- and postsynaptic proteins in adult mouse hippocampal slices. Importantly, this is relevant in two models featuring synaptic deficits. Blocking TWEAK/Fn14 signaling augments synaptic function in hippocampal slices from amyloid-beta-overexpressing mice. After stroke, genetic or pharmacological inhibition of TWEAK/Fn14 signaling augments basal synaptic transmission and normalizes plasticity. Our data support a glial/neuronal axis that critically modifies synaptic physiology and pathophysiology in different contexts in the mature brain and may be a therapeutic target for improving neurophysiological outcomes.
Collapse
Affiliation(s)
- Dávid Nagy
- Clinical Sciences, Biogen, Cambridge, MA 02142
- Biogen Postdoctoral Scientist Program, Cellular Physiology, Biogen, Cambridge, MA 02142
| | - Katelin A Ennis
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Ru Wei
- Chemical Biology and Proteomics, Biogen, Cambridge, MA 02142
| | - Susan C Su
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | | | - Rong-Fang Gu
- Chemical Biology and Proteomics, Biogen, Cambridge, MA 02142
| | - Benbo Gao
- Chemical Biology and Proteomics, Biogen, Cambridge, MA 02142
| | - Ramiro H Massol
- Translational Cellular Sciences, Biogen, Cambridge, MA 02142
| | - Chris Ehrenfels
- Translational Cellular Sciences, Biogen, Cambridge, MA 02142
| | | | - Ankur M Thomas
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Ashley Nelson
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Stefka Gyoneva
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Mihály Hajós
- Clinical Sciences, Biogen, Cambridge, MA 02142
- Comparative Medicine, School of Medicine, Yale University, New Haven, CT 06520
| | - Linda C Burkly
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142;
| |
Collapse
|
17
|
Downregulation of autophagy by 12/15Lipoxygenase worsens the phenotype of an Alzheimer's disease mouse model with plaques, tangles, and memory impairments. Mol Psychiatry 2021; 26:604-613. [PMID: 30279460 DOI: 10.1038/s41380-018-0268-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/03/2018] [Accepted: 09/06/2018] [Indexed: 11/08/2022]
Abstract
Among the different initiating events in Alzheimer's disease (AD) pathogenesis, oxidative stress and neuroinflammation are some of the most iimportant. In the central nervous system, the 12/15Lipoxygenase (12/15LO) enzyme is the source of potent pro-oxidants and inflammatory lipid mediators. Previous works showed that this pathway is up-regulated in AD brains and that its pharmacological targeting modulates the phenotype of transgenic mouse models of the disease. Here we investigate the effect of brain 12/15LO gene delivery on the AD-like phenotype of a mouse model with plaques, tangles and behavioral deficits, the 3xTg mice. Compared with controls, mice over-expressing 12/15LO manifested an exacerbation of spatial learning and memory impairments, which was associated with significant increase in Aβ formation and deposition, and accumulation of hyper-phosphorylated insoluble tau secondary to a down-regulation of autophagy. In addition, the same mice manifested a worsening of neuroinflammation and synaptic pathology. Taken together our study supports the hypothesis that the 12/15LO enzymatic pathway by impairing neuronal autophagy plays a functional role in exacerbating AD-related neuropathologies and cognitive impairments. It provides further critical preclinical evidence to justify developing and testing new and selective 12/15LO inhibitors for AD treatment.
Collapse
|
18
|
Remya C, Dileep KV, Variyar EJ, Zhang KYJ, Omkumar RV, Sadasivan C. Chemical similarity assisted search for acetylcholinesterase inhibitors: Molecular modeling and evaluation of their neuroprotective properties. Int J Biol Macromol 2021; 174:466-476. [PMID: 33497692 DOI: 10.1016/j.ijbiomac.2021.01.148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/07/2021] [Accepted: 01/20/2021] [Indexed: 01/02/2023]
Abstract
Alzheimer's disease (AD) is an obstinate and progressive neurodegenerative disorder, mainly characterized by cognitive decline. Increasing number of AD patients and the lack of promising treatment strategies demands novel therapeutic agents to combat various disease pathologies in AD. Recent progresses in understanding molecular mechanisms in AD helped researchers to streamline the various therapeutic approaches. Inhibiting acetylcholinesterase (AChE) activity has emerged as one of the potential treatment strategies. The present study discusses the identification of two potent AChE inhibitors (ZINC11709541 and ZINC11996936) from ZINC database through conventional in silico approaches and their in vitro validations. These inhibitors have strong preferences towards AChE than butyrylcholinesterase (BChE) and didn't evoke any significant reduction in the cell viability of HEK-293 cells and primary cortical neurons. Furthermore, promising neuroprotective properties has also been displayed against glutamate induced excitotoxicity in primary cortical neurons. The present study proposes two potential drug lead compounds for the treatment of AD, that can be used for further studies and preclinical evaluation.
Collapse
Affiliation(s)
- Chandran Remya
- Department of Biotechnology and Microbiology, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala 670661, India
| | - K V Dileep
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - E Jayadevi Variyar
- Department of Biotechnology and Microbiology, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala 670661, India
| | - Kam Y J Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - R V Omkumar
- Molecular Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud P. O., Trivandrum, Kerala 695014, India
| | - C Sadasivan
- Department of Biotechnology and Microbiology, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala 670661, India.
| |
Collapse
|
19
|
Engin AB, Engin A. Alzheimer's Disease and Protein Kinases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:285-321. [PMID: 33539020 DOI: 10.1007/978-3-030-49844-3_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and accounts for more than 60-80% of all cases of dementia. Loss of pyramidal neurons, extracellular amyloid beta (Abeta) accumulated senile plaques, and neurofibrillary tangles that contain hyperphosphorylated tau constitute the main pathological alterations in AD.Synaptic dysfunction and extrasynaptic N-methyl-D-aspartate receptor (NMDAR) hyperactivation contributes to excitotoxicity in patients with AD. Amyloid precursor protein (APP) and Abeta promoted neurodegeneration develop through the activation of protein kinase signaling cascade in AD. Furthermore, ultimate neuronal death in AD is under control of protein kinases-related signaling pathways. In this chapter, critical check-points within the cross-talk between neuron and protein kinases have been defined regarding the initiation and progression of AD. In this context, amyloid cascade hypothesis, neuroinflammation, oxidative stress, granulovacuolar degeneration, loss of Wnt signaling, Abeta-related synaptic alterations, prolonged calcium ions overload and NMDAR-related synaptotoxicity, damage signals hypothesis and type-3 diabetes are discussed briefly.In addition to clinical perspective of AD pathology, recommendations that might be effective in the treatment of AD patients have been reviewed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
20
|
Jung HY, Kim W, Kwon HJ, Yoo DY, Nam SM, Hahn KR, Yi SS, Choi JH, Kim DW, Yoon YS, Hwang IK. Physical Stress Induced Reduction of Proliferating Cells and Differentiated Neuroblasts Is Ameliorated by Fermented Laminaria japonica Extract Treatment. Mar Drugs 2020; 18:E587. [PMID: 33255381 PMCID: PMC7760277 DOI: 10.3390/md18120587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022] Open
Abstract
Laminaria japonica is widely cultivated in East Asia, including South Korea. Fucoidan, a main component of L. japonica, protects neurons from neurological disorders such as ischemia and traumatic brain injury. In the present study, we examined the effects of extract from fermented L. japonica on the reduction of proliferating cells and neuroblasts in mice that were physically (with electric food shock) or psychologically (with visual, auditory and olfactory sensation) stressed with the help of a communication box. Vehicle (distilled water) or fermented L. japonica extract (50 mg/kg) were orally administered to the mice once a day for 21 days. On the 19th day of the treatment, physical and psychological stress was induced by foot shock using a communication box and thereafter for three days. Plasma corticosterone levels were significantly increased after exposure to physical stress and decreased Ki67 positive proliferating cells and doublecortin immunoreactive neuroblasts. In addition, western blot analysis demonstrated that physical stress as well as psychological stress decreased the expression levels of brain-derived neurotrophic factor (BDNF) and the number of phosphorylated cAMP response element binding protein (pCREB) positive nuclei in the dentate gyrus. Fermentation of L. japonica extract significantly increased the contents of reduced sugar and phenolic compounds. Supplementation with fermented L. japonica extract significantly ameliorated the increases of plasma corticosterone revels and decline in the proliferating cells, neuroblasts, and expression of BDNF and pCREB in the physically stressed mice. These results indicate that fermented L. japonica extract has positive effects in ameliorating the physical stress induced reduction in neurogenesis by modulating BDNF and pCREB expression in the dentate gyrus.
Collapse
Affiliation(s)
- Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - Woosuk Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
- Department of Biomedical Sciences, and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea; (H.J.K.); (D.W.K.)
| | - Dae Young Yoo
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea;
| | - Sung Min Nam
- Department of Anatomy, School of Medicine and Institute for Environmental Science, Wonkwang University, Iksan 54538, Korea;
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - Sun Shin Yi
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Korea;
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea;
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Korea; (H.J.K.); (D.W.K.)
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea; (H.Y.J.); (W.K.); (K.R.H.); (Y.S.Y.)
| |
Collapse
|
21
|
Ghoweri AO, Ouillette L, Frazier HN, Anderson KL, Lin RL, Gant JC, Parent R, Moore S, Murphy GG, Thibault O. Electrophysiological and Imaging Calcium Biomarkers of Aging in Male and Female 5×FAD Mice. J Alzheimers Dis 2020; 78:1419-1438. [PMID: 33164928 PMCID: PMC7836067 DOI: 10.3233/jad-200109] [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] [Indexed: 01/02/2023]
Abstract
BACKGROUND In animal models and tissue preparations, calcium dyshomeostasis is a biomarker of aging and Alzheimer's disease that is associated with synaptic dysfunction, neuritic pruning, and dysregulated cellular processes. It is unclear, however, whether the onset of calcium dysregulation precedes, is concurrent with, or is the product of pathological cellular events (e.g., oxidation, amyloid-β production, and neuroinflammation). Further, neuronal calcium dysregulation is not always present in animal models of amyloidogenesis, questioning its reliability as a disease biomarker. OBJECTIVE Here, we directly tested for the presence of calcium dysregulation in dorsal hippocampal neurons in male and female 5×FAD mice on a C57BL/6 genetic background using sharp electrodes coupled with Oregon-green Bapta-1 imaging. We focused on three ages that coincide with the course of amyloid deposition: 1.5, 4, and 10 months old. METHODS Outcome variables included measures of the afterhyperpolarization, short-term synaptic plasticity, and calcium kinetics during synaptic activation. Quantitative analyses of spatial learning and memory were also conducted using the Morris water maze. Main effects of sex, age, and genotype were identified on measures of electrophysiology and calcium imaging. RESULTS Measures of resting Oregon-green Bapta-1 fluorescence showed significant reductions in the 5×FAD group compared to controls. Deficits in spatial memory, along with increases in Aβ load, were detectable at older ages, allowing us to test for temporal associations with the onset of calcium dysregulation. CONCLUSION Our results provide evidence that reduced, rather than elevated, neuronal calcium is identified in this 5×FAD model and suggests that this surprising result may be a novel biomarker of AD.
Collapse
Affiliation(s)
- Adam O Ghoweri
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Lara Ouillette
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Hilaree N Frazier
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Katie L Anderson
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ruei-Lung Lin
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - John C Gant
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Rachel Parent
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Shannon Moore
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,5037 BSRB, Molecular and Integrative Physiology, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Geoffrey G Murphy
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,5037 BSRB, Molecular and Integrative Physiology, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Olivier Thibault
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
22
|
Li JG, Chiu J, Praticò D. Full recovery of the Alzheimer's disease phenotype by gain of function of vacuolar protein sorting 35. Mol Psychiatry 2020; 25:2630-2640. [PMID: 30733594 PMCID: PMC6685773 DOI: 10.1038/s41380-019-0364-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/14/2018] [Accepted: 01/23/2019] [Indexed: 01/28/2023]
Abstract
Deficit in retromer complex function secondary to lower levels of one of its major components, the vacuolar protein sorting 35 (VPS35), has been reported in Alzheimer's disease (AD) brains. VPS35 genetic reduction results in increased Aβ levels and synaptic pathology in mouse models of the disease. However, whether restoration of its levels has an effect on the AD-like phenotype which includes Aβ plaques, tau tangles and memory impairments remain unknown. In this paper, we investigated the effect of VPS35 gene delivery into the central nervous system on the development of the neuropathology and behavioral deficits of the triple transgenic (3xTg) mice. Compared with controls, animals overexpressing VPS35 had an amelioration of spatial learning and working memory, which associated with a significant reduction in Aβ levels and deposition and tau phosphorylation. Additionally, the same animals had a significant improvement of synaptic pathology and neuroinflammation. In vitro study confirmed that VPS35 up-regulation by reducing total levels of APP and results in a significant decrease in its metabolic products. Our results demonstrate for the first time that VPS35 is directly involved in the development of AD-like phenotype, and for this reason should be considered as a novel therapeutic target for AD.
Collapse
Affiliation(s)
| | | | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
23
|
Reduced Expression of the PP2A Methylesterase, PME-1, or the PP2A Methyltransferase, LCMT-1, Alters Sensitivity to Beta-Amyloid-Induced Cognitive and Electrophysiological Impairments in Mice. J Neurosci 2020; 40:4596-4608. [PMID: 32341098 DOI: 10.1523/jneurosci.2983-19.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/27/2020] [Accepted: 04/17/2020] [Indexed: 12/16/2022] Open
Abstract
Beta-amyloid (Aβ) is thought to play a critical role in Alzheimer's disease (AD), and application of soluble oligomeric forms of Aβ produces AD-like impairments in cognition and synaptic plasticity in experimental systems. We found previously that transgenic overexpression of the PP2A methylesterase, PME-1, or the PP2A methyltransferase, LCMT-1, altered the sensitivity of mice to Aβ-induced impairments, suggesting that PME-1 inhibition may be an effective approach for preventing or treating these impairments. To explore this possibility, we examined the behavioral and electrophysiological effects of acutely applied synthetic Aβ oligomers in male and female mice heterozygous for either a PME-1 KO or an LCMT-1 gene-trap mutation. We found that heterozygous PME-1 KO mice were resistant to Aβ-induced impairments in cognition and synaptic plasticity, whereas LCMT-1 gene-trap mice showed increased sensitivity to Aβ-induced impairments. The heterozygous PME-1 KO mice produced normal levels of endogenous Aβ and exhibited normal electrophysiological responses to picomolar concentrations of Aβ, suggesting that reduced PME-1 expression in these animals protects against Aβ-induced impairments without impacting normal physiological Aβ functions. Together, these data provide additional support for roles for PME-1 and LCMT-1 in regulating sensitivity to Aβ-induced impairments, and suggest that inhibition of PME-1 may constitute a viable therapeutic approach for selectively protecting against the pathologic actions of Aβ in AD.SIGNIFICANCE STATEMENT Elevated levels of β-amyloid (Aβ) in the brain are thought to contribute to the cognitive impairments observed in Alzheimer's disease patients. Here we show that genetically reducing endogenous levels of the PP2A methylesterase, PME-1, prevents the cognitive and electrophysiological impairments caused by acute exposure to pathologic concentrations of Aβ without impairing normal physiological Aβ function or endogenous Aβ production. Conversely, reducing endogenous levels of the PP2A methyltransferase, LCMT-1, increases sensitivity to Aβ-induced impairments. These data offer additional insights into the molecular factors that control sensitivity to Aβ-induced impairments, and suggest that inhibiting PME-1 may constitute a viable therapeutic avenue for preventing Aβ-related impairments in Alzheimer's disease.
Collapse
|
24
|
Vorobyov V, Medvinskaya N, Deev A, Sengpiel F, Bobkova N, Lunin S. Spatial memory deficits initiated by agroclavine injection or olfactory bulbectomy in rats are characterized by different levels of long-term potentiation expression in the hippocampus. Int J Neurosci 2020; 130:1225-1229. [PMID: 32072845 DOI: 10.1080/00207454.2020.1732963] [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: 10/25/2022]
Abstract
Aim: To clarify whether long-term potentiation (LTP) is the mechanism underpinning mnemonic processes. Mathrials and methods: We studied LTP in hippocampal slices from rats whose spatial memory deficit was produced by either olfactory bulbectomy (OBX) or pretreatment with an ergot alkaloid, agroclavine. OBX is accompanied by cholinergic system inhibition whereas agroclavine predominantly activates dopaminergic mediation. The both have been shown to be involved in learning/memory and LTP mechanisms.Results: In OBX- vs. sham-operated rat, we have revealed significant reduction of LTP in hippocampal CA1 region. In contrast, no LTP differences in agroclavine- vs. vehicle-treated rats were observed. Conclusions: These results demonstrate that LTP expression in the hippocampus is dependent on the origin of spatial memory impairment. Furthermore, they suggest that pharmacological and neurodegenerative models of AD might be useful approach for discovery of both AD mechanisms and mixed pathology dementias.
Collapse
Affiliation(s)
- Vasily Vorobyov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Natalia Medvinskaya
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Alexander Deev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Frank Sengpiel
- School of Biosciences and Neuroscience & Mental Health Research Institute, Cardiff University, Museum Avenue, Cardiff, UK
| | - Natalia Bobkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Sergey Lunin
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| |
Collapse
|
25
|
Post-tetanic Potentiation and Depression in Hippocampal Neurons in a Rat Model of Alzheimer’s Disease: Effects of Teucrium Polium Extract. NEUROPHYSIOLOGY+ 2020. [DOI: 10.1007/s11062-020-09827-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
26
|
Effect of Riluzole, a Glutamate Release Inhibitor, on Synaptic Plasticity in the Intrahippocampal Aβ Rat Model of Alzheimer’s Disease. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09820-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
27
|
Bürge M, Kratzer S, Mattusch C, Hofmann C, Kreuzer M, Parsons CG, Rammes G. The anaesthetic xenon partially restores an amyloid beta-induced impairment in murine hippocampal synaptic plasticity. Neuropharmacology 2019; 151:21-32. [PMID: 30940537 DOI: 10.1016/j.neuropharm.2019.03.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/15/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND It is controversially discussed whether general anaesthesia increases the risk of Alzheimer's disease (AD) or accelerates its progression. One important factor in AD pathogenesis is the accumulation of soluble amyloid beta (Aβ) oligomers which affect N-methyl-d-aspartate (NMDA) receptor function and abolish hippocampal long-term potentiation (LTP). NMDA receptor antagonists, at concentrations allowing physiological activation, can prevent Aβ-induced deficits in LTP. The anaesthetics xenon and S-ketamine both act as NMDA receptor antagonists and have been reported to be neuroprotective. In this study, we investigated the effects of subanaesthetic concentrations of these drugs on LTP deficits induced by different Aβ oligomers and compared them to the effects of radiprodil, a NMDA subunit 2B (GluN2B)-selective antagonist. METHODS We applied different Aβ oligomers to murine brain slices and recorded excitatory postsynaptic field potentials before and after high-frequency stimulation in the CA1 region of hippocampus. Radiprodil, xenon and S-ketamine were added and recordings evoked from a second input were measured. RESULTS Xenon and radiprodil, applied at low concentrations, partially restored the LTP deficit induced by pre-incubated Aβ1-42. S-ketamine showed no effect. None of the drugs tested were able to ameliorate Aβ1-40-induced LTP-deficits. CONCLUSIONS Xenon administered at subanaesthetic concentrations partially restored Aβ1-42-induced impairment of LTP, presumably via its weak NMDA receptor antagonism. The effects were in a similar range than those obtained with the NMDA-GluN2B antagonist radiprodil. Our results point to protective properties of xenon in the context of pathological distorted synaptic physiology which might be a meaningful alternative for anaesthesia in AD patients.
Collapse
Affiliation(s)
- Martina Bürge
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany; Department of Perioperative Medicine, Barts Heart Centre, St Bartholomew's Hospital, West Smithfield, London EC1A 7BE, United Kingdom.
| | - Stephan Kratzer
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Corinna Mattusch
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany; Hexal AG, Industriestr. 25, 83607 Holzkirchen, Germany
| | - Carolin Hofmann
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Matthias Kreuzer
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | | | - Gerhard Rammes
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
28
|
Ondrejcak T, Hu NW, Qi Y, Klyubin I, Corbett GT, Fraser G, Perkinton MS, Walsh DM, Billinton A, Rowan MJ. Soluble tau aggregates inhibit synaptic long-term depression and amyloid β-facilitated LTD in vivo. Neurobiol Dis 2019; 127:582-590. [PMID: 30910746 DOI: 10.1016/j.nbd.2019.03.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/01/2019] [Accepted: 03/21/2019] [Indexed: 01/29/2023] Open
Abstract
Soluble synaptotoxic aggregates of the main pathological proteins of Alzheimer's disease, amyloid β-protein (Aß) and tau, have rapid and potent inhibitory effects on long-term potentiation (LTP). Although the promotion of synaptic weakening mechanisms, including long-term depression (LTD), is posited to mediate LTP inhibition by Aß, little is known regarding the action of exogenous tau on LTD. The present study examined the ability of different assemblies of full-length human tau to affect LTD in the dorsal hippocampus of the anaesthetized rat. Unlike Aß, intracerebroventricular injection of soluble aggregates of tau (SτAs), but not monomers or fibrils, potently increased the threshold for LTD induction in a manner that required cellular prion protein. However, MTEP, an antagonist of the putative prion protein coreceptor metabotropic glutamate receptor 5, did not prevent the disruption of synaptic plasticity by SτAs. In contrast, systemic treatment with Ro 25-6981, a selective antagonist at GluN2B subunit-containing NMDA receptors, reduced SτA-mediated inhibition of LTD, but not LTP. Intriguingly, SτAs completely blocked Aß-facilitated LTD, whereas a subthreshold dose of SτAs facilitated Aß-mediated inhibition of LTP. Overall, these findings support the importance of cellular prion protein in mediating a range of, sometimes opposing, actions of soluble Aß and tau aggregates with different effector mechanisms on synaptic plasticity.
Collapse
Affiliation(s)
- Tomas Ondrejcak
- Department of Pharmacology & Therapeutics, Institute of Neuroscience, Trinity College, Dublin 2, Ireland.
| | - Neng-Wei Hu
- Department of Pharmacology & Therapeutics, Institute of Neuroscience, Trinity College, Dublin 2, Ireland; Department of Physiology and Neurobiology, Zhengzhou University School of Medicine, Zhengzhou 450001, China
| | - Yingjie Qi
- Department of Pharmacology & Therapeutics, Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Igor Klyubin
- Department of Pharmacology & Therapeutics, Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Grant T Corbett
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Graham Fraser
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, UK
| | | | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Andrew Billinton
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, UK
| | - Michael J Rowan
- Department of Pharmacology & Therapeutics, Institute of Neuroscience, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
29
|
Ghumatkar PJ, Patil SP, Peshattiwar V, Vijaykumar T, Dighe V, Vanage G, Sathaye S. The modulatory role of phloretin in Aβ 25-35 induced sporadic Alzheimer's disease in rat model. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:327-339. [PMID: 30488341 DOI: 10.1007/s00210-018-1588-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/15/2018] [Indexed: 01/22/2023]
Abstract
Alzheimer's disease (AD) is the leading neurodegenerative disorder with extracellular senile plaques and neurofibrillary tangles as the major hallmarks. The objective was to evaluate the effect of phloretin in a chronic model of sporadic AD by injecting aggregated form of Aβ25-35 peptide sequence intracerebroventricularly (icv) in Wistar rats. To achieve this, male Wistar rats were injected with aggregated Aβ25-35 peptide icv, followed by 21 days phloretin (2.5 mg/kg, 5 mg/kg) administration after recovery period. Barnes maze and elevated plus maze along with the biochemical estimation of antioxidant enzymes activities were conducted. The hippocampus region of the rat brains were stained with Congo red and Nissl stain. TNF-α was estimated in the brain homogenates using the ELISA assay. In this study, phloretin improved the spatial memory formation and retention in Barnes maze test. Additionally, phloretin alleviated the antioxidant defense biomarkers and thereby reduced oxidative stress, decreased TNF-α-mediated neuroinflammation. Furthermore, phloretin treatment showed decreased amyloid beta accumulation in the CA1 region and less number of pyknotic nuclei in the dentate gyrus of the Aβ25-35-injected rat brains. The above experimental findings evinced the promising role of phloretin in Aβ25-35-injected rats and which further envisage its potential to be explored in the treatment of AD.
Collapse
Affiliation(s)
- Priya J Ghumatkar
- Pharmacology Research Laboratory-II, Department of Pharmaceutical Science & Technology, Institute of Chemical Technology (University under Section 3 of UGC Act- 1956, Elite Status & Centre of Excellence-Govt. of Maharashtra, TEQIP Phase II Funded), Matunga (E), Mumbai, Maharashtra, 400019, India
| | - Sachin P Patil
- Pharmacology Research Laboratory-II, Department of Pharmaceutical Science & Technology, Institute of Chemical Technology (University under Section 3 of UGC Act- 1956, Elite Status & Centre of Excellence-Govt. of Maharashtra, TEQIP Phase II Funded), Matunga (E), Mumbai, Maharashtra, 400019, India
| | - Vaibhavi Peshattiwar
- Pharmacology Research Laboratory-II, Department of Pharmaceutical Science & Technology, Institute of Chemical Technology (University under Section 3 of UGC Act- 1956, Elite Status & Centre of Excellence-Govt. of Maharashtra, TEQIP Phase II Funded), Matunga (E), Mumbai, Maharashtra, 400019, India
| | - Tushara Vijaykumar
- National Centre for Preclinical Reproductive and Genetic Toxicology, National Institute for Research in Reproductive Health (NIRRH), Parel, Mumbai, India
| | - Vikas Dighe
- National Centre for Preclinical Reproductive and Genetic Toxicology, National Institute for Research in Reproductive Health (NIRRH), Parel, Mumbai, India
| | - Geeta Vanage
- National Centre for Preclinical Reproductive and Genetic Toxicology, National Institute for Research in Reproductive Health (NIRRH), Parel, Mumbai, India
| | - Sadhana Sathaye
- Pharmacology Research Laboratory-II, Department of Pharmaceutical Science & Technology, Institute of Chemical Technology (University under Section 3 of UGC Act- 1956, Elite Status & Centre of Excellence-Govt. of Maharashtra, TEQIP Phase II Funded), Matunga (E), Mumbai, Maharashtra, 400019, India.
| |
Collapse
|
30
|
Susceptibility to Aβo and TBOA of LTD and Extrasynaptic NMDAR-Dependent Tonic Current in the Aged Rat Hippocampus. Neurochem Res 2018; 44:692-702. [PMID: 30426348 DOI: 10.1007/s11064-018-2677-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 10/26/2018] [Accepted: 11/02/2018] [Indexed: 12/29/2022]
Abstract
Aging, as the major risk factor of Alzheimer's disease (AD), may increase susceptibility to neurodegenerative diseases through many gradual molecular and biochemical changes. Extracellular glutamate homeostasis and extrasynaptic glutamate N-methyl-D-aspartate receptors (NMDAR) are among early synaptic targets of oligomeric amyloid β (Aβo), one of the AD related synaptotoxic protein species. In this study, we asked for the effects of Aβo on long-term depression (LTD), a form of synaptic plasticity dependent on extrasynaptic NMDAR activation, and on a tonic current (TC) resulting from the activation of extrasynaptic NMDAR by ambient glutamate in hippocampal slices from young (3-6-month-old) and aged (24-28-month-old) Sprague-Dawley rats. Aβo significantly enhanced the magnitude of LTD and the amplitude of TC in aged slices compared to young ones. TBOA, a glutamate transporter inhibitor, also significantly increased LTD magnitude and TC amplitude in slices from aged rats, suggesting either an age-related weakness of the glutamate clearance system and/or a facilitated extrasynaptic NMDAR activation. From our present data, we hypothesize that senescence-related impairment of the extrasynaptic environment may be a vector of vulnerability of the aged hippocampus to neurodegenerative promotors such as Aβo.
Collapse
|
31
|
Mechanisms and modulators of cognitive training gain transfer in cognitively healthy aging: study protocol of the AgeGain study. Trials 2018; 19:337. [PMID: 29945638 PMCID: PMC6020358 DOI: 10.1186/s13063-018-2688-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/15/2018] [Indexed: 11/19/2022] Open
Abstract
Background Cognitively healthy older people can increase their performance in cognitive tasks through training. However, training effects are mostly limited to the trained task; thus, training effects only poorly transfer to untrained tasks or other contexts, which contributes to reduced adaptation abilities in aging. Stabilizing transfer capabilities in aging would increase the chance of persistent high performance in activities of daily living including longer independency, and prolonged active participation in social life. The trial AgeGain aims at elaborating the physiological brain mechanisms of transfer in aging and supposed major modulators of transfer capability, especially physical activity, cerebral vascular lesions, and amyloid burden. Methods This 4-year interventional, multicenter, phase 2a cognitive and physical training study will enroll 237 cognitively healthy older subjects in four recruiting centers. The primary endpoint of this trial is the prediction of transfer of cognitive training gains. Secondary endpoints are the structural connectivity of the corpus callosum, Default Mode Network activity, brain-derived neurotrophic factors, motor fitness, and maximal oxygen uptake. Discussion Cognitive transfer allows making use of cognitive training gains in everyday life. Thus, maintenance of transfer capability with aging increases the chance of persistent self-guidance and prolonged active participation in social life, which may support a good quality of life. The AgeGain study aims at identifying older people who will most benefit from cognitive training. It will increase the understanding of the neurobiological mechanisms of transfer in aging and will help in determining the impact of physical activity and sport as well as pathologic factors (such as cerebrovascular disease and amyloid load) on transfer capability. Trial registration German Clinical Trials Register (DRKS), ID: DRKS00013077. Registered on 19 November 2017. Electronic supplementary material The online version of this article (10.1186/s13063-018-2688-2) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Egashira N, Akiyoshi Y, Iba H, Arai T, Hatip-Al-Khatib I, Mishima K, Iwasaki K. Tokishakuyakusan ameliorates spatial memory deficits induced by ovariectomy combined with β-amyloid in rats. J Pharmacol Sci 2018; 136:149-154. [DOI: 10.1016/j.jphs.2018.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/20/2018] [Accepted: 01/26/2018] [Indexed: 12/18/2022] Open
|
33
|
Liu XJ, Wei J, Shang YH, Huang HC, Lao FX. Modulation of AβPP and GSK3β by Endoplasmic Reticulum Stress and Involvement in Alzheimer's Disease. J Alzheimers Dis 2018; 57:1157-1170. [PMID: 28339396 DOI: 10.3233/jad-161111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a dementia disease with neuronal loss and synaptic impairment. This impairment is caused, at least partly, by the generation of two main AD hallmarks, namely the hyperphosphorylated tau protein comprising neurofibrillary tangles and senile plaques containing amyloid-β (Aβ) peptides. The amyloid-β protein precursor (AβPP) and glycogen synthase kinase-3β (GSK3β) are two main proteins associated with AD and are closely correlated with these hallmarks. Recently, both of the proteins were reported to be modulated by endoplasmic reticulum stress (ERS) and are involved in the pathogenesis of AD. The mechanism of ERS plus the modulation of AβPP processing and GSK3β activity by ERS in AD are summarized and explored in this review.
Collapse
Affiliation(s)
- Xin-Jun Liu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Jun Wei
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Ying-Hui Shang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Han-Chang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Feng-Xue Lao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| |
Collapse
|
34
|
A preclinical screen to evaluate pharmacotherapies for the treatment of agitation in dementia. Behav Pharmacol 2018; 28:199-206. [PMID: 28234659 DOI: 10.1097/fbp.0000000000000298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Agitation associated with dementia is frequently reported clinically but has received little attention in preclinical models of dementia. The current study used a 7PA2 CM intracerebroventricular injection model of Alzheimer's disease (AD) to assess acute memory impairment, and a bilateral intrahippocampal (IH) injection model of AD (aggregated Aβ1-42 injections) and a bilateral IH injection model of dementia with Lewy bodies (aggregated NAC61-95 injections) to assess chronic memory impairment in the rat. An alternating-lever cyclic-ratio schedule of operant responding was used for data collection, where incorrect lever perseverations measured executive function (memory) and running response rates (RRR) measured behavioral output (agitation). The results indicate that bilateral IH injections of Aβ1-42 and bilateral IH injections of NAC61-95 decreased memory function and increased RRRs, whereas intracerebroventricular injections of 7PA2 CM decreased memory function but did not increase RRRs. These findings show that using the aggregated peptide IH injection models of dementia to induce chronic neurotoxicity, memory decline was accompanied by elevated behavioral output. This demonstrates that IH peptide injection models of dementia provide a preclinical screen for pharmacological interventions used in the treatment of increased behavioral output (agitation), which also establish detrimental side effects on memory.
Collapse
|
35
|
Corrêa-Velloso JC, Gonçalves MC, Naaldijk Y, Oliveira-Giacomelli Á, Pillat MM, Ulrich H. Pathophysiology in the comorbidity of Bipolar Disorder and Alzheimer's Disease: pharmacological and stem cell approaches. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:34-53. [PMID: 28476640 DOI: 10.1016/j.pnpbp.2017.04.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/28/2017] [Indexed: 12/22/2022]
Abstract
Neuropsychiatric disorders involve various pathological mechanisms, resulting in neurodegeneration and brain atrophy. Neurodevelopmental processes have shown to be critical for the progression of those disorders, which are based on genetic and epigenetic mechanisms as well as on extrinsic factors. We review here common mechanisms underlying the comorbidity of Bipolar Disorders and Alzheimer's Disease, such as aberrant neurogenesis and neurotoxicity, reporting current therapeutic approaches. The understanding of these mechanisms precedes stem cell-based strategies as a new therapeutic possibility for treatment and prevention of Bipolar and Alzheimer's Disease progression. Taking into account the difficulty of studying the molecular basis of disease progression directly in patients, we also discuss the importance of stem cells for effective drug screening, modeling and treating psychiatric diseases, once in vitro differentiation of patient-induced pluripotent stem cells provides relevant information about embryonic origins, intracellular pathways and molecular mechanisms.
Collapse
Affiliation(s)
- Juliana C Corrêa-Velloso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Maria Cb Gonçalves
- Departamento de Neurologia e Neurociências, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, São Paulo, SP 04039-032, Brazil
| | - Yahaira Naaldijk
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Ágatha Oliveira-Giacomelli
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Micheli M Pillat
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
36
|
Yu CC, Wang Y, Shen F, Kong LH, Wang YW, Zhou H, Tang L. High-frequency (50 Hz) electroacupuncture ameliorates cognitive impairment in rats with amyloid beta 1-42-induced Alzheimer's disease. Neural Regen Res 2018; 13:1833-1841. [PMID: 30136700 PMCID: PMC6128060 DOI: 10.4103/1673-5374.238620] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Acupuncture has been shown to ameliorate cognitive impairment of Alzheimer's disease. Acupoints and stimulation frequency influence the therapeutic effect of electroacupuncture. Rat models of Alzheimer's disease were established by injecting amyloid beta 1–42 (Aβ1–42) into the bilateral lateral ventricles. Electroacupuncture at 2, 30, and 50 Hz was carried out at Baihui (GV20; 15° obliquely to a depth of 2 mm) and Shenshu (BL23; perpendicularly to 4–6 mm depth), once a day for 20 minutes (each), for 15 days, taking a break every 7 days. The Morris water maze test was conducted to assess the learning and memory. The expression levels of glycogen synthase kinase-3β (GSK-3β), pSer9-GSK-3β, pTyr216-GSK-3β, amyloid precursor protein and Aβ1–40 in the hippocampus were determined by western blot assay. Results demonstrated that electroacupuncture treatment at different frequencies markedly improved learning and memory ability, increased synaptic curvatures, decreased the width of synaptic clefts, thickened postsynaptic densities, and downregulated the expression of GSK-3β, amyloid precursor protein, and Aβ1–40. pSer9-GSK-3β expression markedly decreased, while pTyr216-GSK-3β expression increased. High-frequency (50 Hz) electroacupuncture was more effective than low (2 Hz) or medium-frequency (30 Hz) electroacupuncture. In conclusion, electroacupuncture treatment exerts a protective effect against Aβ1–42-induced learning and memory deficits and synapse-ultrastructure impairment via inhibition of GSK-3β activity. Moreover, high-frequency electroacupuncture was the most effective therapy.
Collapse
Affiliation(s)
- Chao-Chao Yu
- Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Ying Wang
- Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Feng Shen
- Department of Acupuncture and Moxibustion, College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Li-Hong Kong
- Department of Acupuncture and Moxibustion, College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Ya-Wen Wang
- Department of Acupuncture and Moxibustion, College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Hua Zhou
- Department of Acupuncture and Moxibustion, College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Lei Tang
- Department of Rehabilitation, Wuhan Central Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
37
|
Kashima R, Hata A. The role of TGF-β superfamily signaling in neurological disorders. Acta Biochim Biophys Sin (Shanghai) 2018; 50:106-120. [PMID: 29190314 PMCID: PMC5846707 DOI: 10.1093/abbs/gmx124] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/02/2017] [Indexed: 12/12/2022] Open
Abstract
The TGF-β superfamily signaling is involved in a variety of biological processes during embryogenesis and in adult tissue homeostasis. Faulty regulation of the signaling pathway that transduces the TGF-β superfamily signals accordingly leads to a number of ailments, such as cancer and cardiovascular, metabolic, urinary, intestinal, skeletal, and immune diseases. In recent years, a number of studies have elucidated the essential roles of TGF-βs and BMPs during neuronal development in the maintenance of appropriate innervation and neuronal activity. The new advancement implicates significant roles of the aberrant TGF-β superfamily signaling in the pathogenesis of neurological disorders. In this review, we compile a number of reports implicating the deregulation of TGF-β/BMP signaling pathways in the pathogenesis of cognitive and neurodegenerative disorders in animal models and patients. We apologize in advance that the review falls short of providing details of the role of TGF-β/BMP signaling or mechanisms underlying the pathogenesis of neurological disorders. The goal of this article is to reveal a gap in our knowledge regarding the association between TGF-β/BMP signaling pathways and neuronal tissue homeostasis and development and facilitate the research with a potential to develop new therapies for neurological ailments by modulating the pathways.
Collapse
Affiliation(s)
- Risa Kashima
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA
| | - Akiko Hata
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
38
|
Asam K, Staniszewski A, Zhang H, Melideo SL, Mazzeo A, Voronkov M, Huber KL, Pérez E, Stock M, Stock JB, Arancio O, Nicholls RE. Eicosanoyl-5-hydroxytryptamide (EHT) prevents Alzheimer's disease-related cognitive and electrophysiological impairments in mice exposed to elevated concentrations of oligomeric beta-amyloid. PLoS One 2017; 12:e0189413. [PMID: 29253878 PMCID: PMC5734769 DOI: 10.1371/journal.pone.0189413] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/24/2017] [Indexed: 02/04/2023] Open
Abstract
Soluble forms of oligomeric beta-amyloid (Aβ) are thought to play a central role in Alzheimer's disease (AD). Transgenic manipulation of methylation of the serine/threonine protein phosphatase, PP2A, was recently shown to alter the sensitivity of mice to AD-related impairments resulting from acute exposure to elevated levels of Aβ. In addition, eicosanoyl-5-hydroxytryptamide (EHT), a naturally occurring component from coffee beans that modulates PP2A methylation, was shown to confer therapeutic benefits in rodent models of AD and Parkinson's disease. Here, we tested the hypothesis that EHT protects animals from the pathological effects of exposure to elevated levels of soluble oligomeric Aβ. We treated mice with EHT-containing food at two different doses and assessed the sensitivity of these animals to Aβ-induced behavioral and electrophysiological impairments. We found that EHT administration protected animals from Aβ-induced cognitive impairments in both a radial-arm water maze and contextual fear conditioning task. We also found that both chronic and acute EHT administration prevented Aβ-induced impairments in long-term potentiation. These data add to the accumulating evidence suggesting that interventions with pharmacological agents, such as EHT, that target PP2A activity may be therapeutically beneficial for AD and other neurological conditions.
Collapse
Affiliation(s)
- Kesava Asam
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
| | - Agnieszka Staniszewski
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
| | - Hong Zhang
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
| | - Scott L. Melideo
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Adolfo Mazzeo
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
| | - Michael Voronkov
- Signum Biosciences, 133 Wall Street, Princeton, New Jersey, United States of America
| | - Kristen L. Huber
- Signum Biosciences, 133 Wall Street, Princeton, New Jersey, United States of America
| | - Eduardo Pérez
- Signum Biosciences, 133 Wall Street, Princeton, New Jersey, United States of America
| | - Maxwell Stock
- Signum Biosciences, 133 Wall Street, Princeton, New Jersey, United States of America
| | - Jeffry B. Stock
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- Signum Biosciences, 133 Wall Street, Princeton, New Jersey, United States of America
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
- Department of Medicine, Columbia University, New York, NY, United States of America
| | - Russell E. Nicholls
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
39
|
Brunner D, Flunkert S, Neddens J, Duller S, Scopes D, Treherne J, Hutter-Paier B. SEN1500, a novel oral amyloid-β aggregation inhibitor, attenuates brain pathology in a mouse model of Alzheimer’s disease. Neurosci Lett 2017; 660:96-102. [DOI: 10.1016/j.neulet.2017.09.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 09/01/2017] [Accepted: 09/12/2017] [Indexed: 01/10/2023]
|
40
|
Wilkerson JR, Albanesi JP, Huber KM. Roles for Arc in metabotropic glutamate receptor-dependent LTD and synapse elimination: Implications in health and disease. Semin Cell Dev Biol 2017; 77:51-62. [PMID: 28969983 DOI: 10.1016/j.semcdb.2017.09.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/21/2017] [Accepted: 09/26/2017] [Indexed: 10/18/2022]
Abstract
The Arc gene is robustly transcribed in specific neural ensembles in response to experience-driven activity. Upon induction, Arc mRNA is transported to dendrites, where it can be rapidly and locally translated by activation of metabotropic glutamate receptors (mGluR1/5). mGluR-induced dendritic synthesis of Arc is implicated in weakening or elimination of excitatory synapses by triggering endocytosis of postsynaptic AMPARs in both hippocampal CA1 and cerebellar Purkinje neurons. Importantly, CA1 neurons with experience-induced Arc mRNA are susceptible, or primed for mGluR-induced long-term synaptic depression (mGluR-LTD). Here we review mechanisms and function of Arc in mGluR-LTD and synapse elimination and propose roles for these forms of plasticity in Arc-dependent formation of sparse neural representations of learned experience. We also discuss accumulating evidence linking dysregulation of Arc and mGluR-LTD in human cognitive disorders such as intellectual disability, autism and Alzheimer's disease.
Collapse
Affiliation(s)
- Julia R Wilkerson
- Departments of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Joseph P Albanesi
- Departments of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Kimberly M Huber
- Departments of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| |
Collapse
|
41
|
Chakravarthy M, Chen S, Dodd PR, Veedu RN. Nucleic Acid-Based Theranostics for Tackling Alzheimer's Disease. Theranostics 2017; 7:3933-3947. [PMID: 29109789 PMCID: PMC5667416 DOI: 10.7150/thno.21529] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/28/2017] [Indexed: 02/07/2023] Open
Abstract
Nucleic acid-based technologies have received significant interest in recent years as novel theranostic strategies for various diseases. The approval by the United States Food and Drug Administration (FDA) of Nusinersen, an antisense oligonucleotide drug, for the treatment of spinal muscular dystrophy highlights the potential of nucleic acids to treat neurological diseases, including Alzheimer's disease (AD). AD is a devastating neurodegenerative disease characterized by progressive impairment of cognitive function and behavior. It is the most common form of dementia; it affects more than 20% of people over 65 years of age and leads to death 7-15 years after diagnosis. Intervention with novel agents addressing the underlying molecular causes is critical. Here we provide a comprehensive review on recent developments in nucleic acid-based theranostic strategies to diagnose and treat AD.
Collapse
Affiliation(s)
- Madhuri Chakravarthy
- Centre for Comparative Genomics, Murdoch University, Murdoch, Perth, Australia 6150
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, Perth, Australia 6005
| | - Suxiang Chen
- Centre for Comparative Genomics, Murdoch University, Murdoch, Perth, Australia 6150
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, Perth, Australia 6005
| | - Peter R. Dodd
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Brisbane, Australia 4072
| | - Rakesh N. Veedu
- Centre for Comparative Genomics, Murdoch University, Murdoch, Perth, Australia 6150
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, Perth, Australia 6005
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Brisbane, Australia 4072
| |
Collapse
|
42
|
Liang J, Kulasiri D, Samarasinghe S. Computational investigation of Amyloid-β-induced location- and subunit-specific disturbances of NMDAR at hippocampal dendritic spine in Alzheimer's disease. PLoS One 2017; 12:e0182743. [PMID: 28837653 PMCID: PMC5570373 DOI: 10.1371/journal.pone.0182743] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 07/24/2017] [Indexed: 01/22/2023] Open
Abstract
In Alzheimer’s disease (AD), dysregulation of intracellular Ca2+ signalling has been observed as an early event prior to the presence of clinical symptoms and is believed to be a crucial factor contributing to AD pathogenesis. Amyloid-β oligomers (AβOs) disturb the N-methyl-D-aspartate receptor (NMDAR)-mediated postsynaptic Ca2+ signalling in response to presynaptic stimulation by increasing the availability of extracellular glutamate as well as directly disturbing the NMDARs. The abnormal Ca2+ response can further lead to impairments in long-term potentiation (LTP), an important process in memory formation. In this study, we develop a mathematical model of a CA1 pyramidal dendritic spine and conduct computational experiments. We use this model to mimic alterations by AβOs under AD conditions to investigate how they are involved in the Ca2+ dysregulation in the dendritic spine. The alterations in glutamate availability, as well as NMDAR availability and activity, are studied both individually and globally. The simulation results suggest that alterations in glutamate availability mostly affect the synaptic response and have limited effects on the extrasynaptic receptors. Moreover, overactivation of extrasynaptic NMDARs in AD is unlikely to be induced by presynaptic stimulation, but by upregulation of the resting level of glutamate, possibly resulting from these alterations. Furthermore, internalisation of synaptic NR2A-NMDAR shows greater damage to the postsynaptic Ca2+ response in comparison with the internalisation of NR2B-NMDARs; thus, the suggested neuroprotective role of the latter is very limited during synaptic transmission in AD. We integrate a CaMKII state transition model with the Ca2+ model to further study the effects of alterations of NMDARs in the CaMKII state transition, an important downstream event in the early phase of LTP. The model reveals that cooperation between NR2A- and NR2B-NMDAR is required for LTP induction. Under AD conditions, internalisation of membrane NMDARs is suggested to be the cause of the loss of synapse numbers by disrupting CaMKII-NMDAR formation.
Collapse
Affiliation(s)
- Jingyi Liang
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, New Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, New Zealand
- * E-mail:
| | - Sandhya Samarasinghe
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, New Zealand
| |
Collapse
|
43
|
May LM, Anggono V, Gooch HM, Jang SE, Matusica D, Kerbler GM, Meunier FA, Sah P, Coulson EJ. G-Protein-Coupled Inwardly Rectifying Potassium (GIRK) Channel Activation by the p75 Neurotrophin Receptor Is Required for Amyloid β Toxicity. Front Neurosci 2017; 11:455. [PMID: 28848381 PMCID: PMC5550722 DOI: 10.3389/fnins.2017.00455] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/26/2017] [Indexed: 02/05/2023] Open
Abstract
Alzheimer's disease is characterized by cognitive decline, neuronal degeneration, and the accumulation of amyloid-beta (Aβ). Although, the neurotoxic Aβ peptide is widely believed to trigger neuronal dysfunction and degeneration in Alzheimer's disease, the mechanism by which this occurs is poorly defined. Here we describe a novel, Aβ-triggered apoptotic pathway in which Aβ treatment leads to the upregulation of G-protein activated inwardly rectifying potassium (GIRK/Kir3) channels, causing potassium efflux from neurons and Aβ-mediated apoptosis. Although, GIRK channel activity is required for Aβ-induced neuronal degeneration, we show that it is not sufficient, with coincident signaling by the p75 neurotrophin receptor (p75NTR) also required for potassium efflux and cell death. Our results identify a novel role for GIRK channels in mediating apoptosis, and provide a previously missing mechanistic link between the excitotoxicity of Aβ and its ability to trigger cell death pathways, such as that mediated by p75NTR. We propose that this death-signaling pathway contributes to the dysfunction of neurons in Alzheimer's disease and is responsible for their eventual degeneration.
Collapse
Affiliation(s)
- Linda M May
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia
| | - Victor Anggono
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia.,Clem Jones Centre for Ageing Dementia Research, University of QueenslandBrisbane, QLD, Australia
| | - Helen M Gooch
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia
| | - Se E Jang
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia.,Clem Jones Centre for Ageing Dementia Research, University of QueenslandBrisbane, QLD, Australia
| | - Dusan Matusica
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia.,Centre for Neuroscience, College of Medicine and Public Health, Flinders UniversityAdelaide, SA, Australia
| | - Georg M Kerbler
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia
| | - Frederic A Meunier
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia.,Clem Jones Centre for Ageing Dementia Research, University of QueenslandBrisbane, QLD, Australia
| | - Pankaj Sah
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia
| | - Elizabeth J Coulson
- Queensland Brain Institute, University of QueenslandBrisbane, QLD, Australia.,Clem Jones Centre for Ageing Dementia Research, University of QueenslandBrisbane, QLD, Australia.,School of Biomedical Sciences, University of QueenslandBrisbane, QLD, Australia
| |
Collapse
|
44
|
Gong WY, Wang R, Liu Y, Jin H, Zhao ZW, Wang YL, Li HY, Zhang X, Ni JX. Chronic Monoarthritis Pain Accelerates the Processes of Cognitive Impairment and Increases the NMDAR Subunits NR2B in CA3 of Hippocampus from 5-month-old Transgenic APP/PS1 Mice. Front Aging Neurosci 2017; 9:123. [PMID: 28553223 PMCID: PMC5427068 DOI: 10.3389/fnagi.2017.00123] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/12/2017] [Indexed: 01/09/2023] Open
Abstract
Many factors impact cognitive impairment; however, the effects of chronic pain and the mechanisms underlying these effects on cognitive impairment are currently unknown. Here we tested the hypothesis that chronic pain accelerates the transition from normal cognition to mild cognitive impairment (MCI) in 5-month-old transgenic APP/PS1 mice, an animal model of Alzheimer’s disease (AD), and that neurotoxicity induced by N-methyl-D-aspartic acid receptor (NMDAR) subunits may be involved in this process. Chronic monoarthritis pain was induced in transgenic APP/PS1 mice and 5-month-old wild-type (WT) mice by intra- and pre-articular injections of Freund’s complete adjuvant (FCA) into one knee joint. Pain behavior, learning and memory function, and the distribution and quantity of NMDAR subunits (NR1, NR2A and NR2B) in hippocampal CA1 and CA3 regions were assessed. Our results showed that although persistent and robust monoarthritis pain was induced by the FCA injections, only the transgenic APP/PS1 mice with chronic monoarthritis pain exhibited marked learning and memory impairments. This result suggested that chronic monoarthritis pain accelerated the cognitive impairment process. Furthermore, only transgenic APP/PS1 mice with chronic monoarthritis pain exhibited an overexpression of NR2B and an increased NR2B/NR2A ratio in the hippocampus CA3. These findings suggest that chronic pain is a risk factor for cognitive impairment and that increased neurotoxicity associated with NMDAR subunit activation may underpin the impairment. Thus, NMDARs may be a therapeutic target for the prevention of chronic pain-induced cognitive impairment.
Collapse
Affiliation(s)
- Wei-Yi Gong
- Central Laboratory, Xuanwu Hospital of Capital Medical University, Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain DisordersBeijing, China.,Department of Pain Management, Xuanwu Hospital, Capital Medical UniversityBeijing, China.,Department of Anesthesiology, Fujian Medical University Union HospitalFuzhou, China
| | - Rong Wang
- Central Laboratory, Xuanwu Hospital of Capital Medical University, Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain DisordersBeijing, China
| | - Yuan Liu
- Central Laboratory, Xuanwu Hospital of Capital Medical University, Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain DisordersBeijing, China
| | - He Jin
- Central Laboratory, Xuanwu Hospital of Capital Medical University, Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain DisordersBeijing, China
| | - Zhi-Wei Zhao
- Central Laboratory, Xuanwu Hospital of Capital Medical University, Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain DisordersBeijing, China
| | - Yu-Lan Wang
- Central Laboratory, Xuanwu Hospital of Capital Medical University, Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain DisordersBeijing, China
| | - Hong-Yan Li
- Central Laboratory, Xuanwu Hospital of Capital Medical University, Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain DisordersBeijing, China.,Department of Pain Management, Xuanwu Hospital, Capital Medical UniversityBeijing, China
| | - Xu Zhang
- Central Laboratory, Xuanwu Hospital of Capital Medical University, Laboratory for Neurodegenerative Disease of Ministry of Education, Center of Alzheimer's Disease, Beijing Institute for Brain DisordersBeijing, China
| | - Jia-Xiang Ni
- Department of Pain Management, Xuanwu Hospital, Capital Medical UniversityBeijing, China
| |
Collapse
|
45
|
Santos VV, Stark R, Rial D, Silva HB, Bayliss JA, Lemus MB, Davies JS, Cunha RA, Prediger RD, Andrews ZB. Acyl ghrelin improves cognition, synaptic plasticity deficits and neuroinflammation following amyloid β (Aβ1-40) administration in mice. J Neuroendocrinol 2017; 29. [PMID: 28380673 DOI: 10.1111/jne.12476] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 03/16/2017] [Accepted: 04/03/2017] [Indexed: 12/24/2022]
Abstract
Ghrelin is a metabolic hormone that has neuroprotective actions in a number of neurological conditions, including Parkinson's disease (PD), stroke and traumatic brain injury. Acyl ghrelin treatment in vivo and in vitro also shows protective capacity in Alzheimer's disease (AD). In the present study, we used ghrelin knockout (KO) and their wild-type littermates to test whether or not endogenous ghrelin is protective in a mouse model of AD, in which human amyloid β peptide 1-40 (Aβ1-40 ) was injected into the lateral ventricles i.c.v. Recognition memory, using the novel object recognition task, was significantly impaired in ghrelin KO mice and after i.c.v. Aβ1-40 treatment. These deficits could be prevented by acyl ghrelin injections for 7 days. Spatial orientation, as assessed by the Y-maze task, was also significantly impaired in ghrelin KO mice and after i.c.v. Aβ1-40 treatment. These deficits could be prevented by acyl ghrelin injections for 7 days. Ghrelin KO mice had deficits in olfactory discrimination; however, neither i.c.v. Aβ1-40 treatment, nor acyl ghrelin injections affected olfactory discrimination. We used stereology to show that ghrelin KO and Aβ1-40 increased the total number of glial fibrillary acidic protein expressing astrocytes and ionised calcium-binding adapter expressing microglial in the rostral hippocampus. Finally, Aβ1-40 blocked long-term potentiation induced by high-frequency stimulation and this effect could be acutely blocked with co-administration of acyl ghrelin. Collectively, our studies demonstrate that ghrelin deletion affects memory performance and also that acyl ghrelin treatment may delay the onset of early events of AD. This supports the idea that acyl ghrelin treatment may be therapeutically beneficial with respect to restricting disease progression in AD.
Collapse
Affiliation(s)
- V V Santos
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - R Stark
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - D Rial
- Department of Pharmacology, Center of Biological Sciences, Universidade Federal de Santa Catarina UFSC, Florianópolis, SC, Brazil
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - H B Silva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - J A Bayliss
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - M B Lemus
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - J S Davies
- Molecular Neurobiology, Institute of Life Science, Swansea University, Swansea, UK
| | - R A Cunha
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - R D Prediger
- Department of Pharmacology, Center of Biological Sciences, Universidade Federal de Santa Catarina UFSC, Florianópolis, SC, Brazil
| | - Z B Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
46
|
Vargas-Caballero M, Denk F, Wobst HJ, Arch E, Pegasiou CM, Oliver PL, Shipton OA, Paulsen O, Wade-Martins R. Wild-Type, but Not Mutant N296H, Human Tau Restores Aβ-Mediated Inhibition of LTP in Tau-/- mice. Front Neurosci 2017; 11:201. [PMID: 28484365 PMCID: PMC5401872 DOI: 10.3389/fnins.2017.00201] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 03/24/2017] [Indexed: 12/24/2022] Open
Abstract
Microtubule associated protein tau (MAPT) is involved in the pathogenesis of Alzheimer's disease and many forms of frontotemporal dementia (FTD). We recently reported that Aβ-mediated inhibition of hippocampal long-term potentiation (LTP) in mice requires tau. Here, we asked whether expression of human MAPT can restore Aβ-mediated inhibition on a mouse Tau−/− background and whether human tau with an FTD-causing mutation (N296H) can interfere with Aβ-mediated inhibition of LTP. We used transgenic mouse lines each expressing the full human MAPT locus using bacterial artificial chromosome technology. These lines expressed all six human tau protein isoforms on a Tau−/− background. We found that the human wild-type MAPT H1 locus was able to restore Aβ42-mediated impairment of LTP. In contrast, Aβ42 did not reduce LTP in slices in two independently generated transgenic lines expressing tau protein with the mutation N296H associated with frontotemporal dementia (FTD). Basal phosphorylation of tau measured as the ratio of AT8/Tau5 immunoreactivity was significantly reduced in N296H mutant hippocampal slices. Our data show that human MAPT is able to restore Aβ42-mediated inhibition of LTP in Tau−/− mice. These results provide further evidence that tau protein is central to Aβ-induced LTP impairment and provide a valuable tool for further analysis of the links between Aβ, human tau and impairment of synaptic function.
Collapse
Affiliation(s)
| | - Franziska Denk
- Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK.,Wolfson Centre for Age-Related Diseases, King's College LondonLondon, UK
| | - Heike J Wobst
- Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK.,AstraZeneca-Tufts Lab for Basic and Translational Neuroscience, Tufts University School of MedicineBoston, MA, USA
| | - Emily Arch
- Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Chrysia-Maria Pegasiou
- Biological Sciences and Institute for Life Sciences, University of SouthamptonSouthampton, UK
| | - Peter L Oliver
- Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Olivia A Shipton
- Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK.,Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, UK
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, UK
| | | |
Collapse
|
47
|
Földes A, Kádár K, Kerémi B, Zsembery Á, Gyires K, S Zádori Z, Varga G. Mesenchymal Stem Cells of Dental Origin-Their Potential for Antiinflammatory and Regenerative Actions in Brain and Gut Damage. Curr Neuropharmacol 2017; 14:914-934. [PMID: 26791480 PMCID: PMC5333580 DOI: 10.2174/1570159x14666160121115210] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/14/2015] [Accepted: 01/20/2016] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease, Parkinson’s disease, traumatic brain and spinal cord injury and neuroinflammatory multiple sclerosis are diverse disorders of the central nervous system. However, they are all characterized by various levels of inappropriate inflammatory/immune response along with tissue destruction. In the gastrointestinal system, inflammatory bowel disease (IBD) is also a consequence of tissue destruction resulting from an uncontrolled inflammation. Interestingly, there are many similarities in the immunopathomechanisms of these CNS disorders and the various forms of IBD. Since it is very hard or impossible to cure them by conventional manner, novel therapeutic approaches such as the use of mesenchymal stem cells, are needed. Mesenchymal stem cells have already been isolated from various tissues including the dental pulp and periodontal ligament. Such cells possess transdifferentiating capabilities for different tissue specific cells to serve as new building blocks for regeneration. But more importantly, they are also potent immunomodulators inhibiting proinflammatory processes and stimulating anti-inflammatory mechanisms. The present review was prepared to compare the immunopathomechanisms of the above mentioned neurodegenerative, neurotraumatic and neuroinflammatory diseases with IBD. Additionally, we considered the potential use of mesenchymal stem cells, especially those from dental origin to treat such disorders. We conceive that such efforts will yield considerable advance in treatment options for central and peripheral disorders related to inflammatory degeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gábor Varga
- Departments of Oral Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
48
|
Fonseca R. The aging memory: Modulating epigenetic modifications to improve cognitive function. Neurobiol Learn Mem 2016; 133:182-184. [DOI: 10.1016/j.nlm.2016.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/30/2016] [Accepted: 07/03/2016] [Indexed: 11/16/2022]
|
49
|
Long-term dantrolene treatment reduced intraneuronal amyloid in aged Alzheimer triple transgenic mice. Alzheimer Dis Assoc Disord 2016; 29:184-191. [PMID: 25650693 DOI: 10.1097/wad.0000000000000075] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this study, we investigated the long-term treatment of dantrolene on amyloid and tau neuropathology, brain volume, and cognitive function in aged triple transgenic Alzheimer (3xTg-AD) mice. Fifteen-month old 3xTg-AD mice and wild-type controls were treated with oral dantrolene (5 mg/kg) or vehicle control twice a week for 6 months. Learning and memory were examined using the Morris Water Maze at 21 and 22 months of age. After the behavioral testing, hippocampal and cortical brain volumes were calculated with magnetic resonance imaging and motor function was evaluated using the rotorod. The amyloid burden and tau neurofibrillary tangles in the hippocampus were determined using immunohistochemistry. We found that dantrolene significantly decreased the intraneuronal amyloid accumulation by as much as 76% compared with its corresponding vehicle control, together with a trend to reduce phosphorylated tau in the hippocampus. No significant differences could be detected in hippocampal or cortical brain volume, motor function or cognition among all experimental groups, indicating that the mice were still presymptomatic for Alzheimer disease. Thus, presymptomatic and long-term dantrolene treatment significantly decreased the intraneuronal amyloid burden in aged 3xTg-AD mice before significant changes in brain volume, or cognition.
Collapse
|
50
|
PP2A methylation controls sensitivity and resistance to β-amyloid-induced cognitive and electrophysiological impairments. Proc Natl Acad Sci U S A 2016; 113:3347-52. [PMID: 26951658 DOI: 10.1073/pnas.1521018113] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Elevated levels of the β-amyloid peptide (Aβ) are thought to contribute to cognitive and behavioral impairments observed in Alzheimer's disease (AD). Protein phosphatase 2A (PP2A) participates in multiple molecular pathways implicated in AD, and its expression and activity are reduced in postmortem brains of AD patients. PP2A is regulated by protein methylation, and impaired PP2A methylation is thought to contribute to increased AD risk in hyperhomocysteinemic individuals. To examine further the link between PP2A and AD, we generated transgenic mice that overexpress the PP2A methylesterase, protein phosphatase methylesterase-1 (PME-1), or the PP2A methyltransferase, leucine carboxyl methyltransferase-1 (LCMT-1), and examined the sensitivity of these animals to behavioral and electrophysiological impairments caused by exogenous Aβ exposure. We found that PME-1 overexpression enhanced these impairments, whereas LCMT-1 overexpression protected against Aβ-induced impairments. Neither transgene affected Aβ production or the electrophysiological response to low concentrations of Aβ, suggesting that these manipulations selectively affect the pathological response to elevated Aβ levels. Together these data identify a molecular mechanism linking PP2A to the development of AD-related cognitive impairments that might be therapeutically exploited to target selectively the pathological effects caused by elevated Aβ levels in AD patients.
Collapse
|