51
|
Sharda N, Pengo T, Wang Z, Kandimalla KK. Amyloid-β Peptides Disrupt Interactions Between VAMP-2 and SNAP-25 in Neuronal Cells as Determined by FRET/FLIM. J Alzheimers Dis 2020; 77:423-435. [DOI: 10.3233/jad-200065] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background: Synaptic dysfunction prevalent in Alzheimer’s disease (AD) brain is closely associated with increased accumulation of amyloid-β (Aβ) peptides in the brain parenchyma. It is widely believed that Aβ peptides trigger synaptic dysfunction by interfering with the synaptic vesicular fusion and the release of neurotransmitters, primarily facilitated by the SNARE protein complexes formed by VAMP-2, SNAP-25, and syntaxin-1. However, Aβ interactions with SNARE proteins to ultimately disrupt synaptic vesicular fusion are not well understood. Objective: Our objective is to elucidate mechanisms by which Aβ peptides perturb SNARE complexes. Methods: Intensity (qualitative) and lifetime (quantitative) based measurements involving Forster (fluorescence) resonance energy transfer (FRET) followed by fluorescence lifetime imaging microscopy (FLIM) were employed to investigate the effect of Aβ peptides on dynamic interactions between VAMP-2, labeled with cerulean (Cer) at the N-terminus (FRET donor), and SNAP-25 labeled with citrine (Cit) on the N-terminus (FRET acceptor). The FRET and FLIM interactions at the exocytosis locations on the pre-synaptic membrane were recorded under spontaneous and high potassium evoked conditions. Moreover, cellular accumulation of fluorescein labeled Aβ (F-Aβ) peptides and their co-localization with Cer-VAMP2 was investigated by confocal microscopy. Results: The F-Aβ40 and F-Aβ42 are internalized by differentiated N2A cells, where they colocalize with Cer-VAMP2. Both Aβ40 and Aβ42 decrease interactions between the N-termini of Cer-VAMP2 and Cit-SNAP25 in N2A cells, as determined by FRET/FLIM. Conclusion: By perturbing the N-terminal interactions between VAMP-2 and SNAP-25, Aβ40 and Aβ42, can directly interfere with the SNARE complex formation, which is critical for the docking and fusion of synaptic vesicles.
Collapse
Affiliation(s)
- Nidhi Sharda
- Department of Pharmaceutics and the Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Thomas Pengo
- University of Minnesota Informatics Institute, University Imaging Center, University of Minnesota, Minneapolis, MN, USA
| | - Zengtao Wang
- Department of Pharmaceutics and the Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Karunya K. Kandimalla
- Department of Pharmaceutics and the Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
52
|
Ovsepian SV, Horacek J, O'Leary VB, Hoschl C. The Ups and Downs of BACE1: Walking a Fine Line between Neurocognitive and Other Psychiatric Symptoms of Alzheimer's Disease. Neuroscientist 2020; 27:222-234. [PMID: 32713260 DOI: 10.1177/1073858420940943] [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] [Indexed: 12/14/2022]
Abstract
Although neurocognitive deficit is the best-recognized indicator of Alzheimer's disease (AD), psychotic and other noncognitive symptoms are the prime cause of institutionalization. BACE1 is the rate-limiting enzyme in the production of Aβ of AD, and one of the promising therapeutic targets in countering cognitive decline and amyloid pathology. Changes in BACE1 activity have also emerged to cause significant noncognitive neuropsychiatric symptoms and impairments of circadian rhythms, as evident from clinical trials and reports in transgenic models. In this study, we consider key characteristics of BACE1 with its contribution to neurocognitive deficit and other psychiatric symptoms of AD. We argue that a growing list of noncognitive mental impairments related to pharmacological modulation of BACE1 might present a major obstacle in clinical translation of emerging therapeutic leads targeting this protease. The adverse effects of BACE1 inhibition on mental health call for a revision of treatment strategies that assume indiscriminate inhibition of this key protease, and stress the need for further mechanistic and translational studies.
Collapse
Affiliation(s)
- Saak V Ovsepian
- National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.,International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland
| | - Jiri Horacek
- National Institute of Mental Health, Klecany, Czech Republic
| | - Valerie B O'Leary
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Cyril Hoschl
- National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
53
|
Barthet G, Mulle C. Presynaptic failure in Alzheimer's disease. Prog Neurobiol 2020; 194:101801. [PMID: 32428558 DOI: 10.1016/j.pneurobio.2020.101801] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/24/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022]
Abstract
Synaptic loss is the best correlate of cognitive deficits in Alzheimer's disease (AD). Extensive experimental evidence also indicates alterations of synaptic properties at the early stages of disease progression, before synapse loss and neuronal degeneration. A majority of studies in mouse models of AD have focused on post-synaptic mechanisms, including impairment of long-term plasticity, spine structure and glutamate receptor-mediated transmission. Here we review the literature indicating that the synaptic pathology in AD includes a strong presynaptic component. We describe the evidence indicating presynaptic physiological functions of the major molecular players in AD. These include the amyloid precursor protein (APP) and the two presenilin (PS) paralogs PS1 or PS2, genetically linked to the early-onset form of AD, in addition to tau which accumulates in a pathological form in the AD brain. Three main mechanisms participating in presynaptic functions are highlighted. APP fragments bind to presynaptic receptors (e.g. nAChRs and GABAB receptors), presenilins control Ca2+ homeostasis and Ca2+-sensors, and tau regulates the localization of presynaptic molecules and synaptic vesicles. We then discuss how impairment of these presynaptic physiological functions can explain or forecast the hallmarks of synaptic impairment and associated dysfunction of neuronal circuits in AD. Beyond the physiological roles of the AD-related proteins, studies in AD brains also support preferential presynaptic alteration. This review features presynaptic failure as a strong component of pathological mechanisms in AD.
Collapse
Affiliation(s)
- Gael Barthet
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, University of Bordeaux, France
| | - Christophe Mulle
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, University of Bordeaux, France.
| |
Collapse
|
54
|
Wirths O, Zampar S. Emerging roles of N- and C-terminally truncated Aβ species in Alzheimer’s disease. Expert Opin Ther Targets 2019; 23:991-1004. [DOI: 10.1080/14728222.2019.1702972] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Oliver Wirths
- Department of Psychiatry and Psychotherapy, Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| | - Silvia Zampar
- Department of Psychiatry and Psychotherapy, Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
55
|
Yao KX, Lyu H, Liao MH, Yang L, Gao YP, Liu QB, Wang CK, Lu YM, Jiang GJ, Han F, Wang P. Effect of low-dose Levamlodipine Besylate in the treatment of vascular dementia. Sci Rep 2019; 9:18248. [PMID: 31796756 PMCID: PMC6890753 DOI: 10.1038/s41598-019-47868-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 05/20/2019] [Indexed: 12/20/2022] Open
Abstract
Vascular dementia (VaD) is a complex disorder caused by reduced blood flow in the brain. However, there is no effective pharmacological treatment option available until now. Here, we reported that low-dose levamlodipine besylate could reverse the cognitive impairment in VaD mice model of right unilateral common carotid arteries occlusion (rUCCAO). Oral administration of levamlodipine besylate (0.1 mg/kg) could reduce the latency to find the hidden platform in the MWM test as compared to the vehicle group. Furthermore, vehicle-treated mice revealed reduced phospho-CaMKII (Thr286) levels in the hippocampus, which can be partially restored by levamlodipine besylate (0.1 mg/kg and 0.5 mg/kg) treatment. No significant outcome on microglia and astrocytes were observed following levamlodipine besylate treatment. This data reveal novel findings of the therapeutic potential of low-dose levamlodipine besylate that could considerably enhance the cognitive function in VaD mice.
Collapse
Affiliation(s)
- Kai-Xin Yao
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Hang Lyu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mei-Hua Liao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lin Yang
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Yin-Ping Gao
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Qi-Bing Liu
- School of Pharmacy, Hainan Medical College, Haikou, China
| | - Cheng-Kun Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ying-Mei Lu
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Guo-Jun Jiang
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, Zhejiang, China.
| | - Feng Han
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Ping Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.
| |
Collapse
|
56
|
Baruch-Eliyahu N, Rud V, Braiman A, Priel E. Telomerase increasing compound protects hippocampal neurons from amyloid beta toxicity by enhancing the expression of neurotrophins and plasticity related genes. Sci Rep 2019; 9:18118. [PMID: 31792359 PMCID: PMC6889131 DOI: 10.1038/s41598-019-54741-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 10/03/2019] [Indexed: 12/31/2022] Open
Abstract
The telomerase reverse transcriptase protein, TERT, is expressed in the adult brain and its exogenic expression protects neurons from oxidative stress and from the cytotoxicity of amyloid beta (Aβ). We previously showed that telomerase increasing compounds (AGS) protected neurons from oxidative stress. Therefore, we suggest that increasing TERT by AGS may protect neurons from the Aβ-induced neurotoxicity by influencing genes and factors that participate in neuronal survival and plasticity. Here we used a primary hippocampal cell culture exposed to aggregated Aβ and hippocampi from adult mice. AGS treatment transiently increased TERT gene expression in hippocampal primary cell cultures in the presence or absence of Aβ and protected neurons from Aβ induced neuronal degradation. An increase in the expression of Growth associated protein 43 (GAP43), and Feminizing locus on X-3 genes (NeuN), in the presence or absence of Aβ, and Synaptophysin (SYP) in the presence of Aβ was observed. GAP43, NeuN, SYP, Neurotrophic factors (NGF, BDNF), beta-catenin and cyclin-D1 expression were increased in the hippocampus of AGS treated mice. This data suggests that increasing TERT by pharmaceutical compounds partially exerts its neuroprotective effect by enhancing the expression of neurotrophic factors and neuronal plasticity genes in a mechanism that involved Wnt/beta-catenin pathway.
Collapse
Affiliation(s)
- Natalie Baruch-Eliyahu
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vladislav Rud
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Esther Priel
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
57
|
Abstract
The Amyloid Precursor Protein (APP) is infamous for its proposed pivotal role in the pathogenesis of Alzheimer’s disease (AD). Much research on APP focusses on potential contributions to neurodegeneration, mostly based on mouse models with altered expression or mutated forms of APP. However, cumulative evidence from recent years indicates the indispensability of APP and its metabolites for normal brain physiology. APP contributes to the regulation of synaptic transmission, plasticity, and calcium homeostasis. It plays an important role during development and it exerts neuroprotective effects. Of particular importance is the soluble secreted fragment APPsα which mediates many of its physiological actions, often counteracting the effects of the small APP-derived peptide Aβ. Understanding the contribution of APP for normal functions of the nervous system is of high importance, both from a basic science perspective and also as a basis for generating new pathophysiological concepts and therapeutic approaches in AD. In this article, we review the physiological functions of APP and its metabolites, focusing on synaptic transmission, plasticity, calcium signaling, and neuronal network activity.
Collapse
Affiliation(s)
- Dimitri Hefter
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany.,RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Susann Ludewig
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, Braunschweig, Germany.,Cellular Neurobiology, Zoological Institute, Technical University Braunschweig, Braunschweig, Germany
| | - Andreas Draguhn
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Martin Korte
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, Braunschweig, Germany.,Cellular Neurobiology, Zoological Institute, Technical University Braunschweig, Braunschweig, Germany
| |
Collapse
|
58
|
Gracia L, Lora G, Blair LJ, Jinwal UK. Therapeutic Potential of the Hsp90/Cdc37 Interaction in Neurodegenerative Diseases. Front Neurosci 2019; 13:1263. [PMID: 31824256 PMCID: PMC6882380 DOI: 10.3389/fnins.2019.01263] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/06/2019] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's, Huntington's, and Parkinson's are devastating neurodegenerative diseases that are prevalent in the aging population. Patient care costs continue to rise each year, because there is currently no cure or disease modifying treatments for these diseases. Numerous efforts have been made to understand the molecular interactions governing the disease development. These efforts have revealed that the phosphorylation of proteins by kinases may play a critical role in the aggregation of disease-associated proteins, which is thought to contribute to neurodegeneration. Interestingly, a molecular chaperone complex consisting of the 90 kDa heat shock protein (Hsp90) and Cell Division Cycle 37 (Cdc37) has been shown to regulate the maturation of many of these kinases as well as regulate some disease-associated proteins directly. Thus, the Hsp90/Cdc37 complex may represent a potential drug target for regulating proteins linked to neurodegenerative diseases, through both direct and indirect interactions. Herein, we discuss the broad understanding of many Hsp90/Cdc37 pathways and how this protein complex may be a useful target to regulate the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Liam Gracia
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida-Health, Tampa, FL, United States
| | - Gabriella Lora
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida-Health, Tampa, FL, United States
| | - Laura J. Blair
- Department of Molecular Medicine, Byrd Alzheimer’s Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Umesh K. Jinwal
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida-Health, Tampa, FL, United States
| |
Collapse
|
59
|
Gao Q, Wang Y, Wang X, Fu S, Zhang X, Wang RT, Zhang X. Decreased levels of circulating trimethylamine N-oxide alleviate cognitive and pathological deterioration in transgenic mice: a potential therapeutic approach for Alzheimer's disease. Aging (Albany NY) 2019; 11:8642-8663. [PMID: 31612864 PMCID: PMC6814608 DOI: 10.18632/aging.102352] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/27/2019] [Indexed: 12/23/2022]
Abstract
Trimethylamine-N-oxide (TMAO), a metabolite of gut microbiota, has been implicated in the pathogenesis of Alzheimer’s disease (AD). However, the mechanisms by which TMAO influence cognitive and pathological processes in the AD have not been investigated. In this study, we found that the circulating TMAO levels displayed an age-related increase in both WT and APP/PS1 mice and association with AD-like behavioral and pathological profile. Reduced TMAO by 3,3-Dimethyl-1-butanol (DMB) treatment ameliorated the cognitive deterioration and long-term potentiation (LTP) in APP/PS1 mice. Moreover, DMB treatment also induced a decrease in the Amyloid-β (Aβ)1-42, β-secretase, and β-secretase-cleaved C-terminal fragment (βCTF) levels in the hippocampus. Finally, the effects obtained after treatment with DMB were accompanied by a reduction in circulating clusterin levels and hippocampal neuroinflammatory status in APP/PS1 mice. These findings demonstrate that elevated circulating TMAO during the aging process might deteriorate cognitive function and pathology in APP/PS1 mice.
Collapse
Affiliation(s)
- Qiang Gao
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yuan Wang
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xin Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Shuang Fu
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Xin Zhang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Rui-Tao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Xin Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
60
|
Effect of Different Doses and Times of FK506 on Different Areas of the Hippocampus in the Rat Model of Transient Global Cerebral Ischemia-Reperfusion. Neurol Res Int 2019; 2019:8047672. [PMID: 31467711 PMCID: PMC6699343 DOI: 10.1155/2019/8047672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/26/2019] [Accepted: 07/07/2019] [Indexed: 12/14/2022] Open
Abstract
Background Stroke is a major worldwide problem that is leading to a high mortality rate in humans. Ischemia, as the most common type of stroke, is characterized by tissue damage that can occur due to insufficient blood flow to the brain even for a brief duration, leading to the release of inflammatory factors, cytokines, and free radicals. In this study, we investigated the effective dose and injection time of FK506 as an immunophilin ligand for providing a suitable effect on cells of CA2, CA3, and dentate gyrus of the hippocampus. Methods In this in vivo study, a total of 48 male Wistar rats were divided into nine groups. The ischemia model was induced by the ligation of bilateral common carotid arteries. The doses of FK506 (3, 6, and 10 mg/kg) were administered intravenously (IV) at the beginning of reperfusion, followed by repeated injections (10 mg/kg) at 6, 24, 48, and 72 hours after ischemia, respectively. Brains were removed and prepared for Nissl staining and the TdT-mediated dUTP Nick End Labeling method. Results Data showed that global ischemia did not decrease the number of viable pyramidal cells in CA2 and CA3 regions, but significant differences were observed in the number of viable granular cells and apoptotic bodies in the dentate gyrus between the control and ischemia groups. Repeated doses of 6 mg/kg of FK506 at an interval of 48 hours were deemed to be the suitable dose and best time of injection. Conclusions It seems that FK506 can ameliorate the extent of apoptosis and may be a good candidate for the treatment of ischemia-induced brain damage.
Collapse
|
61
|
Gulisano W, Melone M, Ripoli C, Tropea MR, Li Puma DD, Giunta S, Cocco S, Marcotulli D, Origlia N, Palmeri A, Arancio O, Conti F, Grassi C, Puzzo D. Neuromodulatory Action of Picomolar Extracellular Aβ42 Oligomers on Presynaptic and Postsynaptic Mechanisms Underlying Synaptic Function and Memory. J Neurosci 2019; 39:5986-6000. [PMID: 31127002 PMCID: PMC6650983 DOI: 10.1523/jneurosci.0163-19.2019] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/09/2019] [Accepted: 04/28/2019] [Indexed: 01/01/2023] Open
Abstract
Failure of anti-amyloid-β peptide (Aβ) therapies against Alzheimer's disease (AD), a neurodegenerative disorder characterized by high amounts of the peptide in the brain, raised the question of the physiological role of Aβ released at low concentrations in the healthy brain. To address this question, we studied the presynaptic and postsynaptic mechanisms underlying the neuromodulatory action of picomolar amounts of oligomeric Aβ42 (oAβ42) on synaptic glutamatergic function in male and female mice. We found that 200 pm oAβ42 induces an increase of frequency of miniature EPSCs and a decrease of paired pulse facilitation, associated with an increase in docked vesicle number, indicating that it augments neurotransmitter release at presynaptic level. oAβ42 also produced postsynaptic changes as shown by an increased length of postsynaptic density, accompanied by an increased expression of plasticity-related proteins such as cAMP-responsive element binding protein phosphorylated at Ser133, calcium-calmodulin-dependent kinase II phosphorylated at Thr286, and brain-derived neurotrophic factor, suggesting a role for Aβ in synaptic tagging. These changes resulted in the conversion of early into late long-term potentiation through the nitric oxide/cGMP/protein kinase G intracellular cascade consistent with a cGMP-dependent switch from short- to long-term memory observed in vivo after intrahippocampal administration of picomolar amounts of oAβ42 These effects were present upon extracellular but not intracellular application of the peptide and involved α7 nicotinic acetylcholine receptors. These observations clarified the physiological role of oAβ42 in synaptic function and memory formation providing solid fundamentals for investigating the pathological effects of high Aβ levels in the AD brains.SIGNIFICANCE STATEMENT High levels of oligomeric amyloid-β42 (oAβ42) induce synaptic dysfunction leading to memory impairment in Alzheimer's disease (AD). However, at picomolar concentrations, the peptide is needed to ensure long-term potentiation (LTP) and memory. Here, we show that extracellular 200 pm oAβ42 concentrations increase neurotransmitter release, number of docked vesicles, postsynaptic density length, and expression of plasticity-related proteins leading to the conversion of early LTP into late LTP and of short-term memory into long-term memory. These effects require α7 nicotinic acetylcholine receptors and are mediated through the nitric oxide/cGMP/protein kinase G pathway. The knowledge of Aβ function in the healthy brain might be useful to understand the causes leading to its increase and detrimental effect in AD.
Collapse
Affiliation(s)
- Walter Gulisano
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Marcello Melone
- Section of Neuroscience and Cell Biology, Department Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60020, Italy
- Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), Ancona 60020, Italy
| | - Cristian Ripoli
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome 00168, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Maria Rosaria Tropea
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Domenica D Li Puma
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome 00168, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Salvatore Giunta
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Sara Cocco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Daniele Marcotulli
- Section of Neuroscience and Cell Biology, Department Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60020, Italy
| | - Nicola Origlia
- Neuroscience Institute, Italian National Research Council, Pisa 56100, Italy
| | - Agostino Palmeri
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York 10032
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60020, Italy
- Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), Ancona 60020, Italy
- Foundation for Molecular Medicine, Università Politecnica delle Marche, Ancona 60020, Italy, and
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome 00168, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Daniela Puzzo
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy,
- Oasi Research Institute-IRCCS, Troina, 94018, Italy
| |
Collapse
|
62
|
Amyloid β oligomers suppress excitatory transmitter release via presynaptic depletion of phosphatidylinositol-4,5-bisphosphate. Nat Commun 2019; 10:1193. [PMID: 30867420 PMCID: PMC6416269 DOI: 10.1038/s41467-019-09114-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 02/21/2019] [Indexed: 12/21/2022] Open
Abstract
Amyloid β (Aβ) oligomer-induced aberrant neurotransmitter release is proposed to be a crucial early event leading to synapse dysfunction in Alzheimer's disease (AD). In the present study, we report that the release probability (Pr) at the synapse between the Schaffer collateral (SC) and CA1 pyramidal neurons is significantly reduced at an early stage in mouse models of AD with elevated Aβ production. High nanomolar synthetic oligomeric Aβ42 also suppresses Pr at the SC-CA1 synapse in wild-type mice. This Aβ-induced suppression of Pr is mainly due to an mGluR5-mediated depletion of phosphatidylinositol-4,5-bisphosphate (PIP2) in axons. Selectively inhibiting Aβ-induced PIP2 hydrolysis in the CA3 region of the hippocampus strongly prevents oligomeric Aβ-induced suppression of Pr at the SC-CA1 synapse and rescues synaptic and spatial learning and memory deficits in APP/PS1 mice. These results first reveal the presynaptic mGluR5-PIP2 pathway whereby oligomeric Aβ induces early synaptic deficits in AD.
Collapse
|
63
|
Lanni C, Fagiani F, Racchi M, Preda S, Pascale A, Grilli M, Allegri N, Govoni S. Beta-amyloid short- and long-term synaptic entanglement. Pharmacol Res 2019; 139:243-260. [DOI: 10.1016/j.phrs.2018.11.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/06/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
|
64
|
Marttinen M, Takalo M, Natunen T, Wittrahm R, Gabbouj S, Kemppainen S, Leinonen V, Tanila H, Haapasalo A, Hiltunen M. Molecular Mechanisms of Synaptotoxicity and Neuroinflammation in Alzheimer's Disease. Front Neurosci 2018; 12:963. [PMID: 30618585 PMCID: PMC6301995 DOI: 10.3389/fnins.2018.00963] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/03/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, which is clinically associated with a global cognitive decline and progressive loss of memory and reasoning. According to the prevailing amyloid cascade hypothesis of AD, increased soluble amyloid-β (Aβ) oligomer levels impair the synaptic functions and augment calcium dyshomeostasis, neuroinflammation, oxidative stress as well as the formation of neurofibrillary tangles at specific brain regions. Emerging new findings related to synaptic dysfunction and initial steps of neuroinflammation in AD have been able to delineate the underlying molecular mechanisms, thus reinforcing the development of new treatment strategies and biomarkers for AD beyond the conventional Aβ- and tau-targeted approaches. Particularly, the identification and further characterization of disease-associated microglia and their RNA signatures, AD-associated novel risk genes, neurotoxic astrocytes, and in the involvement of complement-dependent pathway in synaptic pruning and loss in AD have set the outstanding basis for further preclinical and clinical studies. Here, we discuss the recent development and the key findings related to the novel molecular mechanisms and targets underlying the synaptotoxicity and neuroinflammation in AD.
Collapse
Affiliation(s)
- Mikael Marttinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Rebekka Wittrahm
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Sami Gabbouj
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Susanna Kemppainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Ville Leinonen
- Institute of Clinical Medicine – Neurosurgery, University of Eastern Finland, Kuopio, Finland
- Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Unit of Clinical Neuroscience, Neurosurgery, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Heikki Tanila
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Annakaisa Haapasalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
65
|
Vorobyeva AG, Saunders AJ. Amyloid-β interrupts canonical Sonic hedgehog signaling by distorting primary cilia structure. Cilia 2018; 7:5. [PMID: 30140428 PMCID: PMC6098584 DOI: 10.1186/s13630-018-0059-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/08/2018] [Indexed: 12/13/2022] Open
Abstract
Background Primary cilia are small non-motile microtubule and cell membrane protrusions expressed on most vertebrate cells, including cortical and hippocampal neurons. These small organelles serve as sensory structures sampling the extracellular environment and reprogramming the transcriptional machinery in response to environmental change. Primary cilia are decorated with a variety of receptor proteins and are necessary for specific signaling cascades such as the Sonic hedgehog (Shh) pathway. Disrupting cilia structure or function results in a spectrum of diseases collectively referred to as ciliopathies. Common to human ciliopathies is cognitive impairment, a symptom also observed in Alzheimer's disease (AD). One hallmark of AD is accumulation of senile plaques composed of neurotoxic Amyloid-β (Aβ) peptide. The Aβ peptide is generated by the proteolytic cleavage of the amyloid precursor protein (APP). We set out to determine if Aβ affects primary cilia structure and the Shh signaling cascade. Methods We utilized in vitro cell-based assays in combination with fluorescent confocal microscopy to address our study goals. Shh signaling and cilia structure was studied using two different cell lines, mouse NIH3T3 and human HeLa cells. To investigate how Aβ levels affect Shh signaling and cilia structure in these cells, we utilized naturally secreted Aβ as well as synthetic Aβ. Effects on Shh signaling were assessed by luciferase activity while cilia structure was analyzed by fluorescent microscopy. Results Here, we report that APP localizes to primary cilia and Aβ treatment results in distorted primary cilia structure. In addition, we demonstrate that Aβ treatment interrupts canonical Shh signal transduction. Conclusions Overall, our study illustrates that Aβ can alter primary cilia structure suggesting that elevated Aβ levels, like those observed in AD patients, could have similar effects on neuronal primary cilia in the brain. Additionally, our study suggests that Aβ impairs the Shh signaling pathway. Together our findings shed light on two novel targets for future AD therapeutics.
Collapse
|
66
|
Benarroch EE. Glutamatergic synaptic plasticity and dysfunction in Alzheimer disease. Neurology 2018; 91:125-132. [DOI: 10.1212/wnl.0000000000005807] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
67
|
Richter MC, Ludewig S, Winschel A, Abel T, Bold C, Salzburger LR, Klein S, Han K, Weyer SW, Fritz AK, Laube B, Wolfer DP, Buchholz CJ, Korte M, Müller UC. Distinct in vivo roles of secreted APP ectodomain variants APPsα and APPsβ in regulation of spine density, synaptic plasticity, and cognition. EMBO J 2018; 37:embj.201798335. [PMID: 29661886 DOI: 10.15252/embj.201798335] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/23/2018] [Accepted: 03/02/2018] [Indexed: 11/09/2022] Open
Abstract
Increasing evidence suggests that synaptic functions of the amyloid precursor protein (APP), which is key to Alzheimer pathogenesis, may be carried out by its secreted ectodomain (APPs). The specific roles of APPsα and APPsβ fragments, generated by non-amyloidogenic or amyloidogenic APP processing, respectively, remain however unclear. Here, we expressed APPsα or APPsβ in the adult brain of conditional double knockout mice (cDKO) lacking APP and the related APLP2. APPsα efficiently rescued deficits in spine density, synaptic plasticity (LTP and PPF), and spatial reference memory of cDKO mice. In contrast, APPsβ failed to show any detectable effects on synaptic plasticity and spine density. The C-terminal 16 amino acids of APPsα (lacking in APPsβ) proved sufficient to facilitate LTP in a mechanism that depends on functional nicotinic α7-nAChRs. Further, APPsα showed high-affinity, allosteric potentiation of heterologously expressed α7-nAChRs in oocytes. Collectively, we identified α7-nAChRs as a crucial physiological receptor specific for APPsα and show distinct in vivo roles for APPsα versus APPsβ. This implies that reduced levels of APPsα that might occur during Alzheimer pathogenesis cannot be compensated by APPsβ.
Collapse
Affiliation(s)
- Max C Richter
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Susann Ludewig
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, Braunschweig, Germany
| | - Alex Winschel
- Department of Biology, Neurophysiology und Neurosensory Systems, TU Darmstadt, Darmstadt, Germany
| | - Tobias Abel
- Paul-Ehrlich-Institut (PEI), Langen, Germany
| | - Charlotte Bold
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Leonie R Salzburger
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, Braunschweig, Germany
| | - Susanne Klein
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Kang Han
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Sascha W Weyer
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Ann-Kristina Fritz
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Institute of Human Movements Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Bodo Laube
- Department of Biology, Neurophysiology und Neurosensory Systems, TU Darmstadt, Darmstadt, Germany
| | - David P Wolfer
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Institute of Human Movements Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | | | - Martin Korte
- Zoological Institute, Division of Cellular Neurobiology, TU Braunschweig, Braunschweig, Germany.,Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany
| | - Ulrike C Müller
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| |
Collapse
|
68
|
cAMP, cGMP and Amyloid β: Three Ideal Partners for Memory Formation. Trends Neurosci 2018; 41:255-266. [PMID: 29501262 DOI: 10.1016/j.tins.2018.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/25/2018] [Accepted: 02/01/2018] [Indexed: 02/03/2023]
Abstract
cAMP and cGMP are well established second messengers required for long-term potentiation (LTP) and memory formation/consolidation. By contrast, amyloid β (Aβ), mostly known as one of the main culprits for Alzheimer's disease (AD), has received relatively little attention in the context of plasticity and memory. Of note, however, low physiological concentrations of Aβ seem necessary for LTP induction and for memory formation. This should come as no surprise, since hormesis emerged as a central dogma in biology. Additionally, recent evidence indicates that Aβ is one of the downstream effectors for cAMP and cGMP to trigger synaptic plasticity and memory. We argue that these emerging findings depict a new scenario that should change the general view on the amyloidogenic pathway, and that could have significant implications for the understanding of AD and its pharmacological treatment in the future.
Collapse
|
69
|
André EA, Forcelli PA, Pak DT. What goes up must come down: homeostatic synaptic plasticity strategies in neurological disease. FUTURE NEUROLOGY 2018; 13:13-21. [PMID: 29379396 DOI: 10.2217/fnl-2017-0028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/07/2017] [Indexed: 11/21/2022]
Abstract
Brain activity levels are tightly regulated to minimize imbalances in activity state. Deviations from the normal range of activity are deleterious and often associated with neurological disorders. To maintain optimal levels of activity, regulatory mechanisms termed homeostatic synaptic plasticity establish desired 'set points' for neural activity, monitor the network for deviations from the set point and initiate compensatory responses to return activity to the appropriate level that permits physiological function [1,2]. We speculate that impaired homeostatic control may contribute to the etiology of various neurological disorders including epilepsy and Alzheimer's disease, two disorders that exhibit hyperexcitability as a key feature during pathogenesis. Here, we will focus on recent progress in developing homeostatic regulation of neural activity as a therapeutic tool.
Collapse
Affiliation(s)
- Emily A André
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Patrick A Forcelli
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Daniel Ts Pak
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington DC 20057, USA
| |
Collapse
|
70
|
Liu M, Xu Z, Du Z, Wu B, Jin T, Xu K, Xu L, Li E, Xu H. The Identification of Key Genes and Pathways in Glioma by Bioinformatics Analysis. J Immunol Res 2017; 2017:1278081. [PMID: 29362722 PMCID: PMC5736927 DOI: 10.1155/2017/1278081] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 10/16/2017] [Indexed: 02/05/2023] Open
Abstract
Glioma is the most common malignant tumor in the central nervous system. This study aims to explore the potential mechanism and identify gene signatures of glioma. The glioma gene expression profile GSE4290 was analyzed for differentially expressed genes (DEGs). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were applied for the enriched pathways. A protein-protein interaction (PPI) network was constructed to find the hub genes. Survival analysis was conducted to screen and validate critical genes. In this study, 775 downregulated DEGs were identified. GO analysis demonstrated that the DEGs were enriched in cellular protein modification, regulation of cell communication, and regulation of signaling. KEGG analysis indicated that the DEGs were enriched in the MAPK signaling pathway, endocytosis, oxytocin signaling, and calcium signaling. PPI network and module analysis found 12 hub genes, which were enriched in synaptic vesicle cycling rheumatoid arthritis and collecting duct acid secretion. The four key genes CDK17, GNA13, PHF21A, and MTHFD2 were identified in both generation (GSE4412) and validation (GSE4271) dataset, respectively. Regression analysis showed that CDK13, PHF21A, and MTHFD2 were independent predictors. The results suggested that CDK17, GNA13, PHF21A, and MTHFD2 might play important roles and potentially be valuable in the prognosis and treatment of glioma.
Collapse
Affiliation(s)
- Mingfa Liu
- Department of Neurosurgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Zhennan Xu
- Department of Neurosurgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Zepeng Du
- Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Bingli Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Tao Jin
- Department of Neurosurgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Ke Xu
- Department of Neurosurgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| | - Liyan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Haixiong Xu
- Department of Neurosurgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou 515041, China
| |
Collapse
|