1
|
Sollazzo R, Li Puma DD, Aceto G, Paciello F, Colussi C, Vita MG, Giuffrè GM, Pastore F, Casamassa A, Rosati J, Novelli A, Maietta S, Tiziano FD, Marra C, Ripoli C, Grassi C. Structural and functional alterations of neurons derived from sporadic Alzheimer's disease hiPSCs are associated with downregulation of the LIMK1-cofilin axis. Alzheimers Res Ther 2024; 16:267. [PMID: 39702316 DOI: 10.1186/s13195-024-01632-3] [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: 04/11/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by the accumulation of pathological proteins and synaptic dysfunction. This study aims to investigate the molecular and functional differences between human induced pluripotent stem cells (hiPSCs) derived from patients with sporadic AD (sAD) and age-matched controls (healthy subjects, HS), focusing on their neuronal differentiation and synaptic properties in order to better understand the cellular and molecular mechanisms underlying AD pathology. METHODS Skin fibroblasts from sAD patients (n = 5) and HS subjects (n = 5) were reprogrammed into hiPSCs using non-integrating Sendai virus vectors. Through karyotyping, we assessed pluripotency markers (OCT4, SOX2, TRA-1-60) and genomic integrity. Neuronal differentiation was evaluated by immunostaining for MAP2 and NEUN. Electrophysiological properties were measured using whole-cell patch-clamp, while protein expression of Aβ, phosphorylated tau, Synapsin-1, Synaptophysin, PSD95, and GluA1 was quantified by western blot. We then focused on PAK1-LIMK1-Cofilin signaling, which plays a key role in regulating synaptic structure and function, both of which are disrupted in neurodegenerative diseases such as AD. RESULTS sAD and HS hiPSCs displayed similar stemness features and genomic stability. However, they differed in neuronal differentiation and function. sAD-derived neurons (sAD-hNs) displayed increased levels of AD-related proteins, including Aβ and phosphorylated tau. Electrophysiological analyses revealed that while both sAD- and HS-hNs generated action potentials, sAD-hNs exhibited decreased spontaneous synaptic activity. Significant reductions in the expression of synaptic proteins such as Synapsin-1, Synaptophysin, PSD95, and GluA1 were found in sAD-hNs, which are also characterized by reduced neurite length, indicating impaired differentiation. Notably, sAD-hNs demonstrated a marked reduction in LIMK1 phosphorylation, which could be the underlying cause for the changes in cytoskeletal dynamics that we found, leading to the morphological and functional modifications observed in sAD-hNs. To further investigate the involvement of the LIMK1 pathway in the morphological and functional changes observed in sAD neurons, we conducted perturbation experiments using the specific LIMK1 inhibitor, BMS-5. Neurons obtained from healthy subjects treated with the inhibitor showed similar morphological changes to those observed in sAD neurons, confirming that LIMK1 activity is crucial for maintaining normal neuronal structure. Furthermore, administration of the inhibitor to sAD neurons did not exacerbate the morphological alterations, suggesting that LIMK1 activity is already compromised in these cells. CONCLUSION Our findings demonstrate that although sAD- and HS-hiPSCs are similar in their stemness and genomic stability, sAD-hNs exhibit distinct functional and structural anomalies mirroring AD pathology. These anomalies include synaptic dysfunction, altered cytoskeletal organization, and accumulation of AD-related proteins. Our study underscores the usefulness of hiPSCs in modeling AD and provides insights into the disease's molecular underpinnings, thus highlighting potential therapeutic targets.
Collapse
Affiliation(s)
- Raimondo Sollazzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Giuseppe Aceto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Claudia Colussi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Engineering, Istituto Di Analisi Dei Sistemi Ed Informatica "Antonio Ruberti", National Research Council, 00185, Rome, Italy
| | | | | | - Francesco Pastore
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Alessia Casamassa
- Cellular Reprogramming Unit, Fondazione IRCCS Casa, Sollievo Della Sofferenza, 71013 - San Giovanni, Rotondo, Italy
| | - Jessica Rosati
- Cellular Reprogramming Unit, Fondazione IRCCS Casa, Sollievo Della Sofferenza, 71013 - San Giovanni, Rotondo, Italy
- Saint Camillus International, University of Health Sciences, 00131, Rome, Italy
| | - Agnese Novelli
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Sabrina Maietta
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Francesco Danilo Tiziano
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Camillo Marra
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy.
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| |
Collapse
|
2
|
Fonseca-Gomes J, Costa-Coelho T, Ferreira-Manso M, Inteiro-Oliveira S, Vaz SH, Alemãn-Serrano N, Atalaia-Barbacena H, Ribeiro-Rodrigues L, Ramalho RM, Pinto R, Vicente Miranda H, Tanqueiro SR, de Almeida-Borlido C, Ramalho MJ, Miranda-Lourenço C, Belo RF, Ferreira CB, Neves V, Rombo DM, Viais R, Umemori J, Martins IC, Jerónimo-Santos A, Caetano A, Manso N, Mäkinen P, Marttinen M, Takalo M, Bremang M, Pike I, Haapasalo A, Loureiro JA, Pereira MC, Santos NC, Outeiro TF, Castanho MARB, Fernandes A, Hiltunen M, Duarte CB, Castrén E, de Mendonça A, Sebastião AM, Rodrigues TM, Diógenes MJ. A small TAT-TrkB peptide prevents BDNF receptor cleavage and restores synaptic physiology in Alzheimer's disease. Mol Ther 2024; 32:3372-3401. [PMID: 39205389 PMCID: PMC11489560 DOI: 10.1016/j.ymthe.2024.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 08/01/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
In Alzheimer's disease (AD), amyloid β (Aβ)-triggered cleavage of TrkB-FL impairs brain-derived neurotrophic factor (BDNF) signaling, thereby compromising neuronal survival, differentiation, and synaptic transmission and plasticity. Using cerebrospinal fluid and postmortem human brain samples, we show that TrkB-FL cleavage occurs from the early stages of the disease and increases as a function of pathology severity. To explore the therapeutic potential of this disease mechanism, we designed small TAT-fused peptides and screened their ability to prevent TrkB-FL receptor cleavage. Among these, a TAT-TrkB peptide with a lysine-lysine linker prevented TrkB-FL cleavage both in vitro and in vivo and rescued synaptic deficits induced by oligomeric Aβ in hippocampal slices. Furthermore, this TAT-TrkB peptide improved the cognitive performance, ameliorated synaptic plasticity deficits and prevented Tau pathology progression in vivo in the 5XFAD mouse model of AD. No evidence of liver or kidney toxicity was found. We provide proof-of-concept evidence for the efficacy and safety of this therapeutic strategy and anticipate that this TAT-TrkB peptide has the potential to be a disease-modifying drug that can prevent and/or reverse cognitive deficits in patients with AD.
Collapse
Affiliation(s)
- João Fonseca-Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Tiago Costa-Coelho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Mafalda Ferreira-Manso
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Sara Inteiro-Oliveira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Nuno Alemãn-Serrano
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Henrique Atalaia-Barbacena
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Leonor Ribeiro-Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Rita M Ramalho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Rui Pinto
- Laboratory of Systems Integration Pharmacology, Clinical, and Regulatory Science, Research Institute for Medicines (iMED.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal; Dr. Joaquim Chaves Laboratório de Análises Clínicas, 2790-224 Carnaxide, Portugal
| | - Hugo Vicente Miranda
- iNOVA4Health, NOVA Medical School, NMS, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
| | - Sara R Tanqueiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Carolina de Almeida-Borlido
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Maria João Ramalho
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Catarina Miranda-Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Rita F Belo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Catarina B Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Vera Neves
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Diogo M Rombo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Ricardo Viais
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Juzoh Umemori
- Gene and Cell Technology, A.I. Virtanen Institute, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland
| | - Ivo C Martins
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - André Jerónimo-Santos
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - António Caetano
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Nuno Manso
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Petra Mäkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Mikael Marttinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland; Structural and Computational Biology, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Mari Takalo
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Michael Bremang
- Proteome Sciences, Coveham House, Downside Bridge Road, KT11 3EP Cobham, UK
| | - Ian Pike
- Proteome Sciences, Coveham House, Downside Bridge Road, KT11 3EP Cobham, UK
| | - Annakaisa Haapasalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Joana A Loureiro
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Maria Carmo Pereira
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Nuno C Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany; Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany
| | - Miguel A R B Castanho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Adelaide Fernandes
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Mikko Hiltunen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Carlos B Duarte
- CNC - Center for Neuroscience and Cell Biology and Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Eero Castrén
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland
| | - Alexandre de Mendonça
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Tiago M Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal.
| | - Maria José Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal.
| |
Collapse
|
3
|
Jarero-Basulto JJ, Gasca-Martínez Y, Rivera-Cervantes MC, Gasca-Martínez D, Carrillo-González NJ, Beas-Zárate C, Gudiño-Cabrera G. Cytotoxic Effect of Amyloid-β1-42 Oligomers on Endoplasmic Reticulum and Golgi Apparatus Arrangement in SH-SY5Y Neuroblastoma Cells. NEUROSCI 2024; 5:141-157. [PMID: 39483494 PMCID: PMC11469764 DOI: 10.3390/neurosci5020010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 11/03/2024] Open
Abstract
Amyloid-β oligomers are a cytotoxic structure that is key for the establishment of the beginning stages of Alzheimer's disease (AD). These structures promote subcellular alterations that cause synaptic dysfunction, loss of cell communication, and even cell death, generating cognitive deficits. The aim of this study was to investigate the cytotoxic effects of amyloid-β1-42 oligomers (AβOs) on the membranous organelles involved in protein processing: the endoplasmic reticulum (ER) and Golgi apparatus (GA). The results obtained with 10 μM AβOs in SH-SY5Y neuroblastoma cells showed that oligomeric structures are more toxic than monomers because they cause cell viability to decrease as exposure time increases. Survivor cells were analyzed to further understand the toxic effects of AβOs on intracellular organelles. Survivor cells showed morphological alterations associated with abnormal cytoskeleton modification 72-96 h after exposure to AβOs. Moreover, the ER and GA presented rearrangement throughout the cytoplasmic space, which could be attributed to a lack of constitutive protein processing or to previous abnormal cytoskeleton modification. Interestingly, the disorganization of both ER and GA organelles exposed to AβOs is likely an early pathological alteration that could be related to aberrant protein processing and accumulation in AD.
Collapse
Affiliation(s)
- José J Jarero-Basulto
- Cellular Neurobiology Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico; (J.J.J.-B.); (M.C.R.-C.)
| | - Yadira Gasca-Martínez
- Development and Neural Regeneration Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico; (Y.G.-M.); (N.J.C.-G.)
| | - Martha C Rivera-Cervantes
- Cellular Neurobiology Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico; (J.J.J.-B.); (M.C.R.-C.)
| | - Deisy Gasca-Martínez
- Behavioral Analysis Unit, Neurobiology Institute, Campus UNAM, Juriquilla 76230, Mexico;
| | - Nidia Jannette Carrillo-González
- Development and Neural Regeneration Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico; (Y.G.-M.); (N.J.C.-G.)
| | - Carlos Beas-Zárate
- Neurobiotechnology Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico;
| | - Graciela Gudiño-Cabrera
- Development and Neural Regeneration Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico; (Y.G.-M.); (N.J.C.-G.)
| |
Collapse
|
4
|
Sharma C, Mazumder A. A Comprehensive Review on Potential Molecular Drug Targets for the Management of Alzheimer's Disease. Cent Nerv Syst Agents Med Chem 2024; 24:45-56. [PMID: 38305393 DOI: 10.2174/0118715249263300231116062740] [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: 06/20/2023] [Revised: 08/25/2023] [Accepted: 10/04/2023] [Indexed: 02/03/2024]
Abstract
Alzheimer's disease (AD) is an onset and incurable neurodegenerative disorder that has been linked to various genetic, environmental, and lifestyle factors. Recent research has revealed several potential targets for drug development, such as the prevention of Aβ production and removal, prevention of tau hyperphosphorylation, and keeping neurons alive. Drugs that target numerous ADrelated variables have been developed, and early results are encouraging. This review provides a concise map of the different receptor signaling pathways associated with Alzheimer's Disease, as well as insight into drug design based on these pathways. It discusses the molecular mechanisms of AD pathogenesis, such as oxidative stress, aging, Aβ turnover, thiol groups, and mitochondrial activities, and their role in the disease. It also reviews the potential drug targets, in vivo active agents, and docking studies done in AD and provides prospects for future drug development. This review intends to provide more clarity on the molecular processes that occur in Alzheimer's patient's brains, which can be of use in diagnosing and preventing the condition.
Collapse
Affiliation(s)
- Chanchal Sharma
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida-201306, Uttar Pradesh, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida-201306, Uttar Pradesh, India
| |
Collapse
|
5
|
Zhang S, Cao F, Li W, Abumaria N. TRPM7 kinase activity induces amyloid-β degradation to reverse synaptic and cognitive deficits in mouse models of Alzheimer's disease. Sci Signal 2023; 16:eade6325. [PMID: 37433006 DOI: 10.1126/scisignal.ade6325] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 06/15/2023] [Indexed: 07/13/2023]
Abstract
Altered abundance or activity of the dual-function transient receptor potential melastatin-like 7 (TRPM7) protein is implicated in neurodegenerative disorders, including Alzheimer's disease (AD). Toxic aggregation of amyloid-β (Aβ) in neurons is implicated in AD pathology. Here, we found that the kinase activity of TRPM7 is important to stimulate the degradation of Aβ. TRPM7 expression was decreased in hippocampal tissue samples from patients with AD and two mouse models of AD (APP/PS1 and 5XFAD). In cultures of hippocampal neurons from mice, overexpression of full-length TRPM7 or of its functional kinase domain M7CK prevented synapse loss induced by exogenous Aβ. In contrast, this neuroprotection was not afforded by overexpression of either the functional ion channel portion alone or a TRPM7 mutant lacking kinase activity. M7CK overexpression in the hippocampus of young and old 5XFAD mice prevented and reversed, respectively, memory deficits, synapse loss, and Aβ plaque accumulation. In both neurons and mice, M7CK interacted with and activated the metalloprotease MMP14 to promote Aβ degradation. Thus, TRPM7 loss in patients with AD may contribute to the associated Aβ pathology.
Collapse
Affiliation(s)
- Shimeng Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Feifei Cao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Wei Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Nashat Abumaria
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
6
|
Lee AK, Yi N, Khaled H, Feller B, Takahashi H. SorCS1 inhibits amyloid-β binding to neurexin and rescues amyloid-β-induced synaptic pathology. Life Sci Alliance 2023; 6:e202201681. [PMID: 36697254 PMCID: PMC9880023 DOI: 10.26508/lsa.202201681] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Amyloid-β oligomers (AβOs), toxic peptide aggregates found in Alzheimer's disease, cause synapse pathology. AβOs interact with neurexins (NRXs), key synaptic organizers, and this interaction dampens normal trafficking and function of NRXs. Axonal trafficking of NRX is in part regulated by its interaction with SorCS1, a protein sorting receptor, but the impact of SorCS1 regulation of NRXs in Aβ pathology was previously unstudied. Here, we show competition between the SorCS1 ectodomain and AβOs for β-NRX binding and rescue effects of the SorCS1b isoform on AβO-induced synaptic pathology. Like AβOs, the SorCS1 ectodomain binds to NRX1β through the histidine-rich domain of NRX1β, and the SorCS1 ectodomain and AβOs compete for NRX1β binding. In cultured hippocampal neurons, SorCS1b colocalizes with NRX1β on the axon surface, and axonal expression of SorCS1b rescues AβO-induced impairment of NRX-mediated presynaptic organization and presynaptic vesicle recycling and AβO-induced structural defects in excitatory synapses. Thus, our data suggest a role for SorCS1 in the rescue of AβO-induced NRX dysfunction and synaptic pathology, providing the basis for a novel potential therapeutic strategy for Alzheimer's disease.
Collapse
Affiliation(s)
- Alfred Kihoon Lee
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Nayoung Yi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Husam Khaled
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Benjamin Feller
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Hideto Takahashi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
| |
Collapse
|
7
|
Colussi C, Aceto G, Ripoli C, Bertozzi A, Li Puma DD, Paccosi E, D'Ascenzo M, Grassi C. Cytoplasmic HDAC4 recovers synaptic function in the 3×Tg mouse model of Alzheimer's disease. Neuropathol Appl Neurobiol 2023; 49:e12861. [PMID: 36331820 PMCID: PMC10099707 DOI: 10.1111/nan.12861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/26/2022] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
Abstract
AIMS Early dysfunction in Alzheimer's disease (AD) is characterised by alterations of synapse structure and function leading to dysmorphic neurites, decreased spine density, impaired synaptic plasticity and cognitive deficits. The class II member HDAC4, which recently emerged as a crucial factor in shaping synaptic plasticity and memory, was found to be altered in AD. We investigated how the modulation of HDAC4 may contribute to counteracting AD pathogenesis. METHODS Using a cytoplasmic HDAC4 mutant (HDAC4SD ), we studied the recovery of synaptic function in hippocampal tissue and primary neurons from the triple-transgenic mouse model of AD (3×Tg-AD). RESULTS Here, we report that in wild-type mice, HDAC4 is localised at synapses and interacts with postsynaptic proteins, whereas in the 3×Tg-AD, it undergoes nuclear import, reducing its interaction with synaptic proteins. Of note, HDAC4 delocalisation was induced by both amyloid-β and tau accumulation. Overexpression of the HDAC4SD mutant in CA1 pyramidal neurons of organotypic hippocampal slices obtained from 3×Tg-AD mice increased dendritic length and promoted the enrichment of N-cadherin, GluA1, PSD95 and CaMKII proteins at the synaptic level compared with AD neurons transfected with the empty vector. Moreover, HDAC4 overexpression recovered the level of SUMO2/3ylation of PSD95 in AD hippocampal tissue, and in AD organotypic hippocampal slices, the HDAC4SD rescued spine density and synaptic transmission. CONCLUSIONS These results highlight a new role of cytoplasmic HDAC4 in providing a structural and enzymatic regulation of postsynaptic proteins. Our findings suggest that controlling HDAC4 localisation may represent a promising strategy to rescue synaptic function in AD, potentially leading to memory improvement.
Collapse
Affiliation(s)
- Claudia Colussi
- Department of Engineering, Istituto di Analisi dei Sistemi ed Informatica 'Antonio Ruberti', National Research Council, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giuseppe Aceto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alessia Bertozzi
- Department of Engineering, Istituto di Analisi dei Sistemi ed Informatica 'Antonio Ruberti', National Research Council, Rome, Italy
| | - Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Elena Paccosi
- Department of Engineering, Istituto di Analisi dei Sistemi ed Informatica 'Antonio Ruberti', National Research Council, Rome, Italy
| | - Marcello D'Ascenzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
8
|
Puma DDL, Ripoli C, Puliatti G, Pastore F, Lazzarino G, Tavazzi B, Arancio O, Piacentini R, Grassi C. Extracellular tau oligomers affect extracellular glutamate handling by astrocytes through downregulation of GLT-1 expression and impairment of NKA1A2 function. Neuropathol Appl Neurobiol 2022; 48:e12811. [PMID: 35274343 PMCID: PMC9262805 DOI: 10.1111/nan.12811] [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] [Received: 10/08/2021] [Revised: 02/14/2022] [Accepted: 02/26/2022] [Indexed: 11/29/2022]
Abstract
AIMS Several studies reported that astrocytes support neuronal communication by the release of gliotransmitters, including ATP and glutamate. Astrocytes also play a fundamental role in buffering extracellular glutamate in the synaptic cleft, thus limiting the risk of excitotoxicity in neurons. We previously demonstrated that extracellular tau oligomers (ex-oTau), by specifically targeting astrocytes, affect glutamate-dependent synaptic transmission via a reduction in gliotransmitter release. The aim of this work was to determine if ex-oTau also impair the ability of astrocytes to uptake extracellular glutamate, thus further contributing to ex-oTau-dependent neuronal dysfunction. METHODS Primary cultures of astrocytes and organotypic brain slices were exposed to ex-oTau (200 nM) for 1 hour. Extracellular glutamate buffering by astrocytes was studied by: Na+ imaging; electrophysiological recordings; high-performance liquid chromatography; Western blot and immunofluorescence. Experimental paradigms avoiding ex-oTau internalization (i.e., heparin pre-treatment and amyloid precursor protein knockout astrocytes) were used to dissect intracellular vs. extracellular effects of oTau. RESULTS Ex-oTau uploading in astrocytes significantly affected glutamate-transporter-1 expression and function, thus impinging on glutamate buffering activity. Ex-oTau also reduced Na-K-ATPase activity because of pump mislocalization on the plasma membrane, with no significant changes in expression. This effect was independent of oTau internalization and it caused Na+ overload and membrane depolarization in ex-oTau-targeted astrocytes. CONCLUSIONS Ex-oTau exerted a complex action on astrocytes, at both intracellular and extracellular levels. The net effect was dysregulated glutamate signalling in terms of both release and uptake that relied on reduced expression of glutamate-transporter-1, altered function and localization of NKA1A1, and NKA1A2. Consequently, Na+ gradients and all Na+ -dependent transports were affected.
Collapse
Affiliation(s)
- Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulia Puliatti
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Pastore
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giacomo Lazzarino
- UniCamillus Saint Camillus International University of Health Sciences, Rome, Italy
| | - Barbara Tavazzi
- UniCamillus Saint Camillus International University of Health Sciences, Rome, Italy
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and Department of Medicine, Columbia University, New York, NY, United States
| | - Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
9
|
Park H, Hwang Y, Kim J. Transcriptional activation with Cas9 activator nanocomplexes rescues Alzheimer's disease pathology. Biomaterials 2021; 279:121229. [PMID: 34739981 DOI: 10.1016/j.biomaterials.2021.121229] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/02/2021] [Accepted: 10/23/2021] [Indexed: 02/07/2023]
Abstract
CRISPR/Cas9-mediated gene activation is a potential therapeutic strategy that does not induce double-strand break (DSB) DNA damage. However, in vivo gene activation via a Cas9 activator remains a challenge, currently limiting its therapeutic applications. We developed a Cas9 activator nanocomplex that efficiently activates an endogenous gene in the brain in vivo, suggesting its possible application in novel therapeutics. We demonstrated a potential treatment application of the Cas9 activator nanocomplex by activating Adam10 in the mouse brain without introducing insertions and deletions (inDels). Remarkably, in vivo activation of Adam10 with the Cas9 activator nanocomplex improved cognitive deficits in an Alzheimer's disease (AD) mouse model. These results demonstrate the therapeutic potential of Cas9 activator nanocomplexes for a wide range of neurological diseases.
Collapse
Affiliation(s)
- Hanseul Park
- Laboratory of Cell Reprogramming & Gene Editing, Department of Chemistry & Biomedical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Yerim Hwang
- Laboratory of Cell Reprogramming & Gene Editing, Department of Chemistry & Biomedical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Jongpil Kim
- Laboratory of Cell Reprogramming & Gene Editing, Department of Chemistry & Biomedical Engineering, Dongguk University, Seoul, 04620, Republic of Korea.
| |
Collapse
|
10
|
Li Puma DD, Piacentini R, Grassi C. Does Impairment of Adult Neurogenesis Contribute to Pathophysiology of Alzheimer's Disease? A Still Open Question. Front Mol Neurosci 2021; 13:578211. [PMID: 33551741 PMCID: PMC7862134 DOI: 10.3389/fnmol.2020.578211] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Adult hippocampal neurogenesis is a physiological mechanism contributing to hippocampal memory formation. Several studies associated altered hippocampal neurogenesis with aging and Alzheimer's disease (AD). However, whether amyloid-β protein (Aβ)/tau accumulation impairs adult hippocampal neurogenesis and, consequently, the hippocampal circuitry, involved in memory formation, or altered neurogenesis is an epiphenomenon of AD neuropathology contributing negligibly to the AD phenotype, is, especially in humans, still debated. The detrimental effects of Aβ/tau on synaptic function and neuronal viability have been clearly addressed both in in vitro and in vivo experimental models. Until some years ago, studies carried out on in vitro models investigating the action of Aβ/tau on proliferation and differentiation of hippocampal neural stem cells led to contrasting results, mainly due to discrepancies arising from different experimental conditions (e.g., different cellular/animal models, different Aβ and/or tau isoforms, concentrations, and/or aggregation profiles). To date, studies investigating in situ adult hippocampal neurogenesis indicate severe impairment in most of transgenic AD mice; this impairment precedes by several months cognitive dysfunction. Using experimental tools, which only became available in the last few years, research in humans indicated that hippocampal neurogenesis is altered in cognitive declined individuals affected by either mild cognitive impairment or AD as well as in normal cognitive elderly with a significant inverse relationship between the number of newly formed neurons and cognitive impairment. However, despite that such information is available, the question whether impaired neurogenesis contributes to AD pathogenesis or is a mere consequence of Aβ/pTau accumulation is not definitively answered. Herein, we attempted to shed light on this complex and very intriguing topic by reviewing relevant literature on impairment of adult neurogenesis in mouse models of AD and in AD patients analyzing the temporal relationship between the occurrence of altered neurogenesis and the appearance of AD hallmarks and cognitive dysfunctions.
Collapse
Affiliation(s)
- Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
11
|
Tavares MT, Kozikowski AP, Shen S. Mercaptoacetamide: A promising zinc-binding group for the discovery of selective histone deacetylase 6 inhibitors. Eur J Med Chem 2020; 209:112887. [PMID: 33035922 DOI: 10.1016/j.ejmech.2020.112887] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 01/01/2023]
Abstract
Histone deacetylase 6 (HDAC6) is a zinc-dependent HDAC that mainly modulates the acetylation status of non-histone substrates, such as α-tubulin and heat shock protein 90 (HSP90). The activity of HDAC6 plays a critical role in cell proliferation, protein trafficking and degradation, cell shape, migration, as well as regulation of immunomodulatory factors. For this reason, HDAC6 influences the progress of cancers, neurodegenerative disorders, and autoimmune responses. In the last few years, the discovery of selective HDAC6 inhibitors (HDAC6is) has become an attractive research area as five HDAC6is are being investigated in phase I/II clinical trials. However, the hydroxamic acid functional group still represents the predominant zinc-binding group (ZBG), that often suffers from poor pharmacokinetics and mutagenic potential, thus impairing the application of hydroxamate-based HDAC6is for long-term therapies. On the other hand, mercaptoacetamide (MCA)-based HDAC6is comprise a class of compounds that, in some cases, display nanomolar HDAC6 potency and a thousand-fold selectivity over class I HDAC isozymes. Moreover, MCA-based HDAC6is lack the mutagenicity associated with the hydroxamate function and display pharmacological effects, demonstrating the potential of this particular ZBG to improve upon the drug-like properties of HDAC6is. Herein, we summarize for the first time the structure-activity relationships (SARs) of MCA-based HDAC6is, discuss their HDAC6 selectivity at the molecular level using inhibitor-HDAC co-crystal structures, and further provide our perspective regarding their drug metabolism, pharmacokinetics, and pharmacological properties.
Collapse
Affiliation(s)
- Maurício T Tavares
- Department of Molecular Medicine, Scripps Research, Jupiter, FL, 33458, United States
| | | | - Sida Shen
- Departments of Chemistry, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, And Center for Developmental Therapeutics, Northwestern University, Evanston, IL 60208, United States.
| |
Collapse
|
12
|
Ripoli C, Spinelli M, Natale F, Fusco S, Grassi C. Glucose Overload Inhibits Glutamatergic Synaptic Transmission: A Novel Role for CREB-Mediated Regulation of Synaptotagmins 2 and 4. Front Cell Dev Biol 2020; 8:810. [PMID: 32974347 PMCID: PMC7466440 DOI: 10.3389/fcell.2020.00810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/31/2020] [Indexed: 01/02/2023] Open
Abstract
Glucose metabolism derangement is critically involved in the age-related memory loss but the underlying molecular mechanisms are still poorly understood. In a mouse model of type 1 diabetes we found memory impairment associated with inhibition of the transcription factor CREB and alteration of pre- and post-synaptic protein expression in the hippocampus. Accordingly, glucose excess negatively affected activity-dependent CREB phosphorylation and CREB-mediated mRNA expression of synaptic proteins in hippocampal primary neurons. Specifically, glucose excess inhibited the activity-dependent recruitment of CREB on the regulatory sequences of synaptotagmin (SYT) 2 and 4 promoters and the expression of SYT4 protein. As a result, high glucose affected both the frequency of miniature excitatory postsynaptic currents and NMDA receptor-mediated currents in autaptic hippocampal neuronal cultures. Collectively, our findings highlight novel mechanisms underlying hyperglycaemia-related memory loss, including CREB-dependent downregulation of synaptotagmin expression.
Collapse
Affiliation(s)
- Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Matteo Spinelli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesca Natale
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
13
|
Bekkers JM. Autaptic Cultures: Methods and Applications. Front Synaptic Neurosci 2020; 12:18. [PMID: 32425765 PMCID: PMC7203343 DOI: 10.3389/fnsyn.2020.00018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/01/2020] [Indexed: 11/13/2022] Open
Abstract
Neurons typically form daisy chains of synaptic connections with other neurons, but they can also form synapses with themselves. Although such self-synapses, or autapses, are comparatively rare in vivo, they are surprisingly common in dissociated neuronal cultures. At first glance, autapses in culture seem like a mere curiosity. However, by providing a simple model system in which a single recording electrode gives simultaneous access to the pre- and postsynaptic compartments, autaptic cultures have proven to be invaluable in facilitating important and elegant experiments in the area of synaptic neuroscience. Here, I provide detailed protocols for preparing and recording from autaptic cultures (also called micro-island or microdot cultures). Variations on the basic procedure are presented, as well as practical tips for optimizing the outcomes. I also illustrate the utility of autaptic cultures by reviewing the types of experiments that have used them over the past three decades. These examples serve to highlight the power and elegance of this simple model system, and will hopefully inspire new experiments for the interrogation of synaptic function.
Collapse
Affiliation(s)
- John M Bekkers
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
14
|
Li Puma DD, Piacentini R, Leone L, Gironi K, Marcocci ME, De Chiara G, Palamara AT, Grassi C. Herpes Simplex Virus Type-1 Infection Impairs Adult Hippocampal Neurogenesis via Amyloid-β Protein Accumulation. Stem Cells 2019; 37:1467-1480. [PMID: 31381841 DOI: 10.1002/stem.3072] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/21/2019] [Indexed: 12/14/2022]
Abstract
We previously reported that Herpes simplex virus type-1 (HSV-1) infection of cultured neurons triggered intracellular accumulation of amyloid-β protein (Aβ) markedly impinging on neuronal functions. Here, we demonstrated that HSV-1 affects in vitro and in vivo adult hippocampal neurogenesis by reducing neural stem/progenitor cell (NSC) proliferation and their neuronal differentiation via intracellular Aβ accumulation. Specifically, cultured NSCs were more permissive for HSV-1 replication than mature neurons and, once infected, they exhibited reduced proliferation (assessed by 5'-bromo-deoxyuridine incorporation, Ki67 immunoreactivity, and Sox2 mRNA expression) and impaired neuronal differentiation in favor of glial phenotype (evaluated by immunoreactivity for the neuronal marker MAP2, the glial marker glial fibrillary astrocyte protein, and the expression of the proneuronal genes Mash1 and NeuroD1). Similarly, impaired adult neurogenesis was observed in the subgranular zone of hippocampal dentate gyrus of an in vivo model of recurrent HSV-1 infections, that we recently set up and characterized, with respect to mock-infected mice. The effects of HSV-1 on neurogenesis did not depend on cell death and were due to Aβ accumulation in infected NSCs. Indeed, they were: (a) reverted, in vitro, by the presence of either β/γ-secretase inhibitors preventing Aβ production or the specific 4G8 antibody counteracting the action of intracellular Aβ; (b) not detectable, in vivo, in HSV-1-infected amyloid precursor protein knockout mice, unable to produce and accumulate Aβ. Given the critical role played by adult neurogenesis in hippocampal-dependent memory and learning, our results suggest that multiple virus reactivations in the brain may contribute to Alzheimer's disease phenotype by also targeting NSCs. Stem Cells 2019;37:1467-1480.
Collapse
Affiliation(s)
- Domenica Donatella Li Puma
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Roberto Piacentini
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lucia Leone
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Katia Gironi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Elena Marcocci
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Giovanna De Chiara
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy.,San Raffaele Pisana, IRCCS, Telematic University, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
15
|
Fattorini G, Ripoli C, Cocco S, Spinelli M, Mattera A, Grassi C, Conti F. Glutamate/GABA co-release selectively influences postsynaptic glutamate receptors in mouse cortical neurons. Neuropharmacology 2019; 161:107737. [PMID: 31398382 DOI: 10.1016/j.neuropharm.2019.107737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 07/24/2019] [Accepted: 08/06/2019] [Indexed: 02/08/2023]
Abstract
Cultured rat cortical neurons co-expressing VGLUT1 and VGAT (mixed synapses) co-release Glu and GABA. Here, mixed synapses were studied in cultured mouse cortical neurons to verify whether in mice mixed synapses co-release Glu and GABA, and to gain insight into how they may influence excitation/inhibition balance. Results showed the existence of synapses and autapses that co-release Glu and GABA in cultured mouse cortical neurons, and the ability of both neurotransmitters to evoke postsynaptic responses mediated by ionotropic receptors. We studied the short-term plasticity of glutamatergic, GABAergic, and mixed responses and we found that the kinetics of mixPSC amplitude depression was similar to that observed in EPSCs, but it was different from that of IPSCs. We found similar presynaptic release characteristics in glutamatergic and mixed synapses. Analysis of postsynaptic features, obtained by measuring AMPAR- and NMDAR-mediated currents, showed that AMPAR-mediated currents were significantly higher in pure glutamatergic than in mixed synapses, whereas NMDAR-mediated currents were not significantly different from those measured in mixed synapses. Overall, our findings demonstrate that glutamatergic and mixed synapses share similar electrophysiological properties. However, co-release of GABA and Glu influences postsynaptic ionotropic glutamatergic receptor subtypes, thus selectively influencing AMPAR-mediated currents. These findings strengthen the view that mixed neurons can play a key role in CNS development and in maintaining the excitation-inhibition balance.
Collapse
Affiliation(s)
- Giorgia Fattorini
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, 60026, Ancona, Italy; Center for Neurobiology of Aging, INRCA, IRCCS, Ancona, Italy.
| | - Cristian Ripoli
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli - IRCCS, 00168, Rome, Italy
| | - Sara Cocco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Matteo Spinelli
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Andrea Mattera
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli - IRCCS, 00168, Rome, Italy
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, 60026, Ancona, Italy; Center for Neurobiology of Aging, INRCA, IRCCS, Ancona, Italy
| |
Collapse
|
16
|
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: 67] [Impact Index Per Article: 11.2] [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
|
17
|
Pilkington AW, Donohoe GC, Akhmedov NG, Ferrebee T, Valentine SJ, Legleiter J. Hydrogen Peroxide Modifies Aβ-Membrane Interactions with Implications for Aβ 40 Aggregation. Biochemistry 2019; 58:2893-2905. [PMID: 31187978 DOI: 10.1021/acs.biochem.9b00233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is pathologically characterized by the formation of extracellular senile plaques, predominately comprised of aggregated β-amyloid (Aβ), deposited in the brain. Aβ aggregation can result in a myriad of distinct aggregate species, from soluble oligomers to insoluble fibrils. Aβ strongly interacts with membranes, which can be linked to a variety of potential toxic mechanisms associated with AD. Oxidative damage accompanies the formation of Aβ aggregates, with a 10-50% proportion of Aβ aggregates being oxidized in vivo. Hydrogen peroxide (H2O2) is a reactive oxygen species implicated in a number of neurodegenerative diseases. Recent evidence has demonstrated that the H2O2 concentration fluctuates rapidly in the brain, resulting in large concentration spikes, especially in the synaptic cleft. Here, the impact of environmental H2O2 on Aβ aggregation in the presence and absence of lipid membranes is investigated. Aβ40 was exposed to H2O2, resulting in the selective oxidation of methionine 35 (Met35) to produce Aβ40Met35[O]. While oxidation mildly reduced the rate of Aβ aggregation and produced a distinct fibril morphology at high H2O2 concentrations, H2O2 had a much more pronounced impact on Aβ aggregation in the presence of total brain lipid extract vesicles. The impact of H2O2 on Aβ aggregation in the presence of lipids was associated with a reduced affinity of Aβ for the vesicle surface. However, this reduced vesicle affinity was predominately associated with lipid peroxidation rather than Aβ oxidation.
Collapse
Affiliation(s)
- Albert W Pilkington
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States
| | - Gregory C Donohoe
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States
| | - Novruz G Akhmedov
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States
| | - Timothy Ferrebee
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States
| | - Stephen J Valentine
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States
| | - Justin Legleiter
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States.,Blanchette Rockefeller Neurosciences Institutes , West Virginia University , 1 Medical Center Drive , P.O. Box 9303, Morgantown , West Virginia 26505 , United States.,Department of Neuroscience , West Virginia University , 1 Medical Center Drive , P.O. Box 9303, Morgantown , West Virginia 26505 , United States
| |
Collapse
|
18
|
Gavello D, Calorio C, Franchino C, Cesano F, Carabelli V, Carbone E, Marcantoni A. Early Alterations of Hippocampal Neuronal Firing Induced by Abeta42. Cereb Cortex 2019; 28:433-446. [PMID: 27999123 DOI: 10.1093/cercor/bhw377] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Indexed: 12/11/2022] Open
Abstract
We studied the effect of Amyloid β 1-42 oligomers (Abeta42) on Ca2+ dependent excitability profile of hippocampal neurons. Abeta42 is one of the Amyloid beta peptides produced by the proteolytic processing of the amyloid precursor protein and participates in the initiating event triggering the progressive dismantling of synapses and neuronal circuits. Our experiments on cultured hippocampal network reveal that Abeta42 increases intracellular Ca2+ concentration by 46% and inhibits firing discharge by 19%. More precisely, Abeta42 differently regulates ryanodine (RyRs), NMDA receptors (NMDARs), and voltage gated calcium channels (VGCCs) by increasing Ca2+ release through RyRs and inhibiting Ca2+ influx through NMDARs and VGCCs. The overall increased intracellular Ca2+ concentration causes stimulation of K+ current carried by big conductance Ca2+ activated potassium (BK) channels and hippocampal network firing inhibition. We conclude that Abeta42 alters neuronal function by means of at least 4 main targets: RyRs, NMDARs, VGCCs, and BK channels. The development of selective modulators of these channels may in turn be useful for developing effective therapies that could enhance the quality of life of AD patients during the early onset of the pathology.
Collapse
Affiliation(s)
- Daniela Gavello
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| | - Chiara Calorio
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| | - Claudio Franchino
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| | - Federico Cesano
- Department of Chemistry Via Pietro Giuria 7, Torino University, 10125 Torino, Italy
| | - Valentina Carabelli
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| | - Emilio Carbone
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| | - Andrea Marcantoni
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| |
Collapse
|
19
|
Latulippe J, Lotito D, Murby D. A mathematical model for the effects of amyloid beta on intracellular calcium. PLoS One 2018; 13:e0202503. [PMID: 30133494 PMCID: PMC6105003 DOI: 10.1371/journal.pone.0202503] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 08/03/2018] [Indexed: 12/21/2022] Open
Abstract
The accumulation of Alzheimer's disease (AD) associated Amyloid beta (Aβ) oligomers can trigger aberrant intracellular calcium (Ca2+) levels by disrupting the intrinsic Ca2+ regulatory mechanism within cells. These disruptions can cause changes in homeostasis levels that can have detrimental effects on cell function and survival. Although studies have shown that Aβ can interfere with various Ca2+ fluxes, the complexity of these interactions remains elusive. We have constructed a mathematical model that simulates Ca2+ patterns under the influence of Aβ. Our simulations shows that Aβ can increase regions of mixed-mode oscillations leading to aberrant signals under various conditions. We investigate how Aβ affects individual flux contributions through inositol triphosphate (IP3) receptors, ryanodine receptors, and membrane pores. We demonstrate that controlling for the ryanodine receptor's maximal kinetic reaction rate may provide a biophysical way of managing aberrant Ca2+ signals. The influence of a dynamic model for IP3 production is also investigated under various conditions as well as the impact of changes in membrane potential. Our model is one of the first to investigate the effects of Aβ on a variety of cellular mechanisms providing a base modeling scheme from which further studies can draw on to better understand Ca2+ regulation in an AD environment.
Collapse
Affiliation(s)
- Joe Latulippe
- Mathematics Department, Norwich University, Northfield, Vermont, United States of America
- * E-mail:
| | - Derek Lotito
- Chemistry and Biochemistry Department, Norwich University, Northfield, Vermont, United States of America
| | - Donovan Murby
- Mathematics Department, Norwich University, Northfield, Vermont, United States of America
| |
Collapse
|
20
|
Biliverdin Reductase-A Mediates the Beneficial Effects of Intranasal Insulin in Alzheimer Disease. Mol Neurobiol 2018; 56:2922-2943. [PMID: 30073505 DOI: 10.1007/s12035-018-1231-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/10/2018] [Indexed: 12/19/2022]
Abstract
Impairment of biliverdin reductase-A (BVR-A) is an early event leading to brain insulin resistance in AD. Intranasal insulin (INI) administration is under evaluation as a strategy to alleviate brain insulin resistance; however, the molecular mechanisms underlying INI beneficial effects are still unclear. We show that INI improves insulin signaling activation in the hippocampus and cortex of adult and aged 3×Tg-AD mice by ameliorating BVR-A activation. These changes were associated with a reduction of nitrosative stress, Tau phosphorylation, and Aβ oligomers in brain, along with improved cognitive functions. The role of BVR-A was strengthened by showing that cells lacking BVR-A: (i) develop insulin resistance if treated with insulin and (ii) can be recovered from insulin resistance only if treated with a BVR-A-mimetic peptide. These novel findings shed light on the mechanisms underlying INI treatment effects and suggest BVR-A as potential therapeutic target to prevent brain insulin resistance in AD.
Collapse
|
21
|
Gulisano W, Melone M, Li Puma DD, Tropea MR, Palmeri A, Arancio O, Grassi C, Conti F, Puzzo D. The effect of amyloid-β peptide on synaptic plasticity and memory is influenced by different isoforms, concentrations, and aggregation status. Neurobiol Aging 2018; 71:51-60. [PMID: 30092511 DOI: 10.1016/j.neurobiolaging.2018.06.025] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/28/2018] [Accepted: 06/19/2018] [Indexed: 01/06/2023]
Abstract
The increase of oligomeric amyloid-beta (oAβ) has been related to synaptic dysfunction, thought to be the earliest event in Alzheimer's disease pathophysiology. Conversely, the suppression of endogenous Aβ impaired synaptic plasticity and memory, suggesting that the peptide is needed in the healthy brain. However, different species, aggregation forms and concentrations of Aβ might differently influence synaptic function/dysfunction. Here, we have tested the contribution of monomeric and oligomeric Aβ42 and Aβ40 at 200 nM and 200 pM concentrations on hippocampal long-term potentiation and spatial memory. We found that, when at 200 nM, oAβ40, oAβ42, and monomeric Aβ42 impaired long-term potentiation and memory, whereas only oAβ42 200 pM enhanced synaptic plasticity and memory and rescued the detrimental effect due to depletion of endogenous Aβ. Interestingly, quantification of monomer-like and oligomer-like species carried out by transmission electron microscopy revealed an increase of the monomer/oligomer ratio in the oAβ42 200 pM preparation, suggesting that the content of monomers and oligomers depends on the final concentration of the solution.
Collapse
Affiliation(s)
- Walter Gulisano
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Marcello Melone
- Department Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy; Center for Neurobiology of Aging, INRCA IRCCS, Ancona, Italy
| | - Domenica D Li Puma
- Institute of Human Physiology, Università Cattolica Medical School, Rome, Italy
| | - Maria Rosaria Tropea
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Agostino Palmeri
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica Medical School, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Fiorenzo Conti
- Department Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy; Center for Neurobiology of Aging, INRCA IRCCS, Ancona, Italy; Foundation for Molecular Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Daniela Puzzo
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
22
|
Gulisano W, Maugeri D, Baltrons MA, Fà M, Amato A, Palmeri A, D’Adamio L, Grassi C, Devanand D, Honig LS, Puzzo D, Arancio O. Role of Amyloid-β and Tau Proteins in Alzheimer's Disease: Confuting the Amyloid Cascade. J Alzheimers Dis 2018; 64:S611-S631. [PMID: 29865055 PMCID: PMC8371153 DOI: 10.3233/jad-179935] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The "Amyloid Cascade Hypothesis" has dominated the Alzheimer's disease (AD) field in the last 25 years. It posits that the increase of amyloid-β (Aβ) is the key event in AD that triggers tau pathology followed by neuronal death and eventually, the disease. However, therapeutic approaches aimed at decreasing Aβ levels have so far failed, and tau-based clinical trials have not yet produced positive findings. This begs the question of whether the hypothesis is correct. Here we have examined literature on the role of Aβ and tau in synaptic dysfunction, memory loss, and seeding and spreading of AD, highlighting important parallelisms between the two proteins in all of these phenomena. We discuss novel findings showing binding of both Aβ and tau oligomers to amyloid-β protein precursor (AβPP), and the requirement for the presence of this protein for both Aβ and tau to enter neurons and induce abnormal synaptic function and memory. Most importantly, we propose a novel view of AD pathogenesis in which extracellular oligomers of Aβ and tau act in parallel and upstream of AβPP. Such a view will call for a reconsideration of therapeutic approaches directed against Aβ and tau, paving the way to an increased interest toward AβPP, both for understanding the pathogenesis of the disease and elaborating new therapeutic strategies.
Collapse
Affiliation(s)
- Walter Gulisano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Daniele Maugeri
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Marian A. Baltrons
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Biochemistry and Molecular Biology and Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Mauro Fà
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Arianna Amato
- Department of Anaesthesiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Agostino Palmeri
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Luciano D’Adamio
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University, Newark, NJ, USA
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - D.P. Devanand
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Lawrence S. Honig
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| |
Collapse
|
23
|
Brain insulin resistance impairs hippocampal synaptic plasticity and memory by increasing GluA1 palmitoylation through FoxO3a. Nat Commun 2017; 8:2009. [PMID: 29222408 PMCID: PMC5722929 DOI: 10.1038/s41467-017-02221-9] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 11/13/2017] [Indexed: 11/08/2022] Open
Abstract
High-fat diet (HFD) and metabolic diseases cause detrimental effects on hippocampal synaptic plasticity, learning, and memory through molecular mechanisms still poorly understood. Here, we demonstrate that HFD increases palmitic acid deposition in the hippocampus and induces hippocampal insulin resistance leading to FoxO3a-mediated overexpression of the palmitoyltransferase zDHHC3. The excess of palmitic acid along with higher zDHHC3 levels causes hyper-palmitoylation of AMPA glutamate receptor subunit GluA1, hindering its activity-dependent trafficking to the plasma membrane. Accordingly, AMPAR current amplitudes and, more importantly, their potentiation underlying synaptic plasticity were inhibited, as well as hippocampal-dependent memory. Hippocampus-specific silencing of Zdhhc3 and, interestingly enough, intranasal injection of the palmitoyltransferase inhibitor, 2-bromopalmitate, counteract GluA1 hyper-palmitoylation and restore synaptic plasticity and memory in HFD mice. Our data reveal a key role of FoxO3a/Zdhhc3/GluA1 axis in the HFD-dependent impairment of cognitive function and identify a novel mechanism underlying the cross talk between metabolic and cognitive disorders.
Collapse
|
24
|
Identification of post-translational modifications of Aβ peptide in platelet membranes from patients with cerebral amyloid angiopathy. J Neurol Sci 2017; 383:11-17. [DOI: 10.1016/j.jns.2017.08.3269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 01/13/2023]
|
25
|
Human Brain-Derived Aβ Oligomers Bind to Synapses and Disrupt Synaptic Activity in a Manner That Requires APP. J Neurosci 2017; 37:11947-11966. [PMID: 29101243 DOI: 10.1523/jneurosci.2009-17.2017] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/19/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022] Open
Abstract
Compelling genetic evidence links the amyloid precursor protein (APP) to Alzheimer's disease (AD) and several theories have been advanced to explain the relationship. A leading hypothesis proposes that a small amphipathic fragment of APP, the amyloid β-protein (Aβ), self-associates to form soluble aggregates that impair synaptic and network activity. Here, we used the most disease-relevant form of Aβ, protein isolated from AD brain. Using this material, we show that the synaptotoxic effects of Aβ depend on expression of APP and that the Aβ-mediated impairment of synaptic plasticity is accompanied by presynaptic effects that disrupt the excitatory/inhibitory (E/I) balance. The net increase in the E/I ratio and inhibition of plasticity are associated with Aβ localizing to synapses and binding of soluble Aβ aggregates to synapses requires the expression of APP. Our findings indicate a role for APP in AD pathogenesis beyond the generation of Aβ and suggest modulation of APP expression as a therapy for AD.SIGNIFICANCE STATEMENT Here, we report on the plasticity-disrupting effects of amyloid β-protein (Aβ) isolated from Alzheimer's disease (AD) brain and the requirement of amyloid precursor protein (APP) for these effects. We show that Aβ-containing AD brain extracts block hippocampal LTP, augment glutamate release probability, and disrupt the excitatory/inhibitory balance. These effects are associated with Aβ localizing to synapses and genetic ablation of APP prevents both Aβ binding and Aβ-mediated synaptic dysfunctions. Our results emphasize the importance of APP in AD and should stimulate new studies to elucidate APP-related targets suitable for pharmacological manipulation.
Collapse
|
26
|
Piacentini R, Puma DDL, Mainardi M, Lazzarino G, Tavazzi B, Arancio O, Grassi C. Reduced gliotransmitter release from astrocytes mediates tau-induced synaptic dysfunction in cultured hippocampal neurons. Glia 2017; 65:1302-1316. [PMID: 28519902 PMCID: PMC5520670 DOI: 10.1002/glia.23163] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 01/19/2023]
Abstract
Tau is a microtubule-associated protein exerting several physiological functions in neurons. In Alzheimer's disease (AD) misfolded tau accumulates intraneuronally and leads to axonal degeneration. However, tau has also been found in the extracellular medium. Recent studies indicated that extracellular tau uploaded from neurons causes synaptic dysfunction and contributes to tau pathology propagation. Here we report novel evidence that extracellular tau oligomers are abundantly and rapidly accumulated in astrocytes where they disrupt intracellular Ca2+ signaling and Ca2+ -dependent release of gliotransmitters, especially ATP. Consequently, synaptic vesicle release, the expression of pre- and postsynaptic proteins, and mEPSC frequency and amplitude were reduced in neighboring neurons. Notably, we found that tau uploading from astrocytes required the amyloid precursor protein, APP. Collectively, our findings suggests that astrocytes play a critical role in the synaptotoxic effects of tau via reduced gliotransmitter availability, and that astrocytes are major determinants of tau pathology in AD.
Collapse
Affiliation(s)
- Roberto Piacentini
- Institute of Human Physiology, Medical School, Università Cattolica, Largo F. Vito 1, 00168, Rome, Italy
| | - Domenica Donatella Li Puma
- Institute of Human Physiology, Medical School, Università Cattolica, Largo F. Vito 1, 00168, Rome, Italy
| | - Marco Mainardi
- Institute of Human Physiology, Medical School, Università Cattolica, Largo F. Vito 1, 00168, Rome, Italy
| | - Giacomo Lazzarino
- Institute of Biochemistry and Clinical Biochemistry, Medical School, Università Cattolica, Largo F. Vito 1, 00168, Rome, Italy
| | - Barbara Tavazzi
- Institute of Biochemistry and Clinical Biochemistry, Medical School, Università Cattolica, Largo F. Vito 1, 00168, Rome, Italy
| | - Ottavio Arancio
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, 630 W 168th St., NY 10032 USA
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Università Cattolica, Largo F. Vito 1, 00168, Rome, Italy
| |
Collapse
|
27
|
Mainardi M, Spinelli M, Scala F, Mattera A, Fusco S, D'Ascenzo M, Grassi C. Loss of Leptin-Induced Modulation of Hippocampal Synaptic Trasmission and Signal Transduction in High-Fat Diet-Fed Mice. Front Cell Neurosci 2017; 11:225. [PMID: 28804449 PMCID: PMC5532388 DOI: 10.3389/fncel.2017.00225] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/14/2017] [Indexed: 12/16/2022] Open
Abstract
Hippocampal plasticity is triggered by a variety of stimuli including sensory inputs, neurotrophins and inflammation. Leptin, whose primary function is to regulate food intake and energy expenditure, has been recently shown to affect hippocampal neurogenesis and plasticity. Interestingly, mice fed a high-fat diet (HFD) exhibit impaired hippocampal function, but the underlying mechanisms are poorly understood. To address this issue, we compared leptin responsiveness of hippocampal neurons in control and HFD-fed mice by combining single-cell electrophysiology and biochemical assays. We found that leptin modulated spontaneous and evoked synaptic transmission in control, but not HFD, mice. This functional impairment was paralleled by blunted activation of STAT-3, one of the key signal transduction pathways controlled by the fully functional isoform of the leptin receptor, ObRb. In addition, SOCS-3 expression was non-responsive to leptin, indicating that modulation of negative feedback impinging on ObRb was also altered. Our results advance the understanding of leptin action on hippocampal plasticity and, more importantly, suggest that leptin resistance is a key determinant of hippocampal dysfunction associated with hypercaloric diet.
Collapse
Affiliation(s)
- Marco Mainardi
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Matteo Spinelli
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Federico Scala
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Andrea Mattera
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Salvatore Fusco
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Marcello D'Ascenzo
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| |
Collapse
|
28
|
Puzzo D, Piacentini R, Fá M, Gulisano W, Li Puma DD, Staniszewski A, Zhang H, Tropea MR, Cocco S, Palmeri A, Fraser P, D'Adamio L, Grassi C, Arancio O. LTP and memory impairment caused by extracellular Aβ and Tau oligomers is APP-dependent. eLife 2017; 6. [PMID: 28696204 PMCID: PMC5529106 DOI: 10.7554/elife.26991] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/10/2017] [Indexed: 12/21/2022] Open
Abstract
The concurrent application of subtoxic doses of soluble oligomeric forms of human amyloid-beta (oAβ) and Tau (oTau) proteins impairs memory and its electrophysiological surrogate long-term potentiation (LTP), effects that may be mediated by intra-neuronal oligomers uptake. Intrigued by these findings, we investigated whether oAβ and oTau share a common mechanism when they impair memory and LTP in mice. We found that as already shown for oAβ, also oTau can bind to amyloid precursor protein (APP). Moreover, efficient intra-neuronal uptake of oAβ and oTau requires expression of APP. Finally, the toxic effect of both extracellular oAβ and oTau on memory and LTP is dependent upon APP since APP-KO mice were resistant to oAβ- and oTau-induced defects in spatial/associative memory and LTP. Thus, APP might serve as a common therapeutic target against Alzheimer's Disease (AD) and a host of other neurodegenerative diseases characterized by abnormal levels of Aβ and/or Tau. DOI:http://dx.doi.org/10.7554/eLife.26991.001
Collapse
Affiliation(s)
- Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Roberto Piacentini
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mauro Fá
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, United States
| | - Walter Gulisano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Domenica D Li Puma
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Agnes Staniszewski
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, United States
| | - Hong Zhang
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, United States
| | - Maria Rosaria Tropea
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sara Cocco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Agostino Palmeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Paul Fraser
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Luciano D'Adamio
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, United States
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ottavio Arancio
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New york, United States
| |
Collapse
|
29
|
Marsh J, Bagol SH, Williams RSB, Dickson G, Alifragis P. Synapsin I phosphorylation is dysregulated by beta-amyloid oligomers and restored by valproic acid. Neurobiol Dis 2017. [PMID: 28647556 DOI: 10.1016/j.nbd.2017.06.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease is the most prevalent form of dementia in the elderly but the precise causal mechanisms are still not fully understood. Growing evidence supports a significant role for Aβ42 oligomers in the development and progression of Alzheimer's. For example, intracellular soluble Aβ oligomers are thought to contribute to the early synaptic dysfunction associated with Alzheimer's disease, but the molecular mechanisms underlying this effect are still unclear. Here, we identify a novel mechanism that contributes to our understanding of the reported synaptic dysfunction. Using primary rat hippocampal neurons exposed for a short period of time to Aβ42 oligomers, we show a disruption in the activity-dependent phosphorylation cycle of SynapsinI at Ser9. SynapsinI is a pre-synaptic protein that responds to neuronal activity and regulates the availability of synaptic vesicles to participate in neurotransmitter release. Phosphorylation of SynapsinI at Ser9, modulates its distribution and interaction with synaptic vesicles. Our results show that in neurons exposed to Aβ42 oligomers, the levels of phosphorylated Ser9 of SynapsinI remain elevated during the recovery period following neuronal activity. We then investigated if this effect could be targeted by a putative therapeutic regime using valproic acid (a short branch-chained fatty acid) that has been proposed as a treatment for Alzheimer's disease. Exposure of Aβ42 treated neurons to valproic acid, showed that it restores the physiological regulation of SynapsinI after depolarisation. Our data provide a new insight on Aβ42-mediated pathology in Alzheimer's disease and supports the use of Valproic acid as a possible pharmaceutical intervention for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Jade Marsh
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Saifuddien Haji Bagol
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Robin S B Williams
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - George Dickson
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Pavlos Alifragis
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK.
| |
Collapse
|
30
|
Ripoli C. Engrampigenetics: Epigenetics of engram memory cells. Behav Brain Res 2017; 325:297-302. [DOI: 10.1016/j.bbr.2016.11.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/24/2016] [Accepted: 11/26/2016] [Indexed: 12/21/2022]
|
31
|
Amyloid-β Oligomers Interact with Neurexin and Diminish Neurexin-mediated Excitatory Presynaptic Organization. Sci Rep 2017; 7:42548. [PMID: 28211900 PMCID: PMC5304201 DOI: 10.1038/srep42548] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/12/2017] [Indexed: 01/29/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by excessive production and deposition of amyloid-beta (Aβ) proteins as well as synapse dysfunction and loss. While soluble Aβ oligomers (AβOs) have deleterious effects on synapse function and reduce synapse number, the underlying molecular mechanisms are not well understood. Here we screened synaptic organizer proteins for cell-surface interaction with AβOs and identified a novel interaction between neurexins (NRXs) and AβOs. AβOs bind to NRXs via the N-terminal histidine-rich domain (HRD) of β-NRX1/2/3 and alternatively-spliced inserts at splicing site 4 of NRX1/2. In artificial synapse-formation assays, AβOs diminish excitatory presynaptic differentiation induced by NRX-interacting proteins including neuroligin1/2 (NLG1/2) and the leucine-rich repeat transmembrane protein LRRTM2. Although AβOs do not interfere with the binding of NRX1β to NLG1 or LRRTM2, time-lapse imaging revealed that AβO treatment reduces surface expression of NRX1β on axons and that this reduction depends on the NRX1β HRD. In transgenic mice expressing mutated human amyloid precursor protein, synaptic expression of β-NRXs, but not α-NRXs, decreases. Thus our data indicate that AβOs interact with NRXs and that this interaction inhibits NRX-mediated presynaptic differentiation by reducing surface expression of axonal β-NRXs, providing molecular and mechanistic insights into how AβOs lead to synaptic pathology in AD.
Collapse
|
32
|
|
33
|
Song JM, Sung YM, Nam JH, Yoon H, Chung A, Moffat E, Jung M, Pak DTS, Kim J, Hoe HS. A Mercaptoacetamide-Based Class II Histone Deacetylase Inhibitor Increases Dendritic Spine Density via RasGRF1/ERK Pathway. J Alzheimers Dis 2016; 51:591-604. [PMID: 26890742 DOI: 10.3233/jad-150717] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The accumulation of amyloid-β (Aβ) leads to the loss of dendritic spines and synapses, which is hypothesized to cause cognitive impairments in Alzheimer's disease (AD) patients. In our previous study, we demonstrated that a novel mercaptoacetamide-based class II histone deacetylase inhibitor (HDACI), known as W2, decreased Aβ levels and improved learning and memory in mice. However, the underlying mechanism of this effect is unknown. OBJECTIVE Because dendritic spine formation is associated with cognitive performance, here we investigated whether HDACI W2 regulates dendritic spine density and its molecular mechanism of action. METHODS To examine the effect of HDACI W2 on dendritic spine density, we conducted morphological analysis of dendritic spines using GFP transfection and Golgi staining. In addition, to determine the molecular mechanism of W2 effects on spines, we measured the levels of mRNAs and proteins involved in the Ras signaling pathway using quantitative real-time PCR, immunocytochemistry, and western analysis. RESULTS We found that HDACI W2 altered dendritic spine density and morphology in vitro and in vivo. Additionally, W2 increased the mRNA or protein levels of Ras GRF1 and phospho-ERK. Moreover, knockdown of RasGRF1 and inhibition of ERK activity prevented the W2-mediated spinogenesis in primary hippocampal neurons. CONCLUSION Our Class II-selective HDACI W2 promotes the formation and growth of dendritic spines in a RasGRF1 and ERK dependent manner in primary hippocampal neurons.
Collapse
Affiliation(s)
- Jung Min Song
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - You Me Sung
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Jin Han Nam
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, Korea
| | - Hyejin Yoon
- Department of Neuroscience, Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Andrew Chung
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Emily Moffat
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Mira Jung
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC, USA
| | - Jungsu Kim
- Department of Neuroscience, Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Hyang-Sook Hoe
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA.,Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Dong-gu, Daegu, Korea
| |
Collapse
|
34
|
Lee K, Kim H, An K, Kwon OB, Park S, Cha JH, Kim MH, Lee Y, Kim JH, Cho K, Kim HS. Replenishment of microRNA-188-5p restores the synaptic and cognitive deficits in 5XFAD Mouse Model of Alzheimer's Disease. Sci Rep 2016; 6:34433. [PMID: 27708404 PMCID: PMC5052619 DOI: 10.1038/srep34433] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/12/2016] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs have emerged as key factors in development, neurogenesis and synaptic functions in the central nervous system. In the present study, we investigated a pathophysiological significance of microRNA-188-5p (miR-188-5p) in Alzheimer's disease (AD). We found that oligomeric Aβ1-42 treatment diminished miR-188-5p expression in primary hippocampal neuron cultures and that miR-188-5p rescued the Aβ1-42-mediated synapse elimination and synaptic dysfunctions. Moreover, the impairments in cognitive function and synaptic transmission observed in 7-month-old five familial AD (5XFAD) transgenic mice, were ameliorated via viral-mediated expression of miR-188-5p. miR-188-5p expression was down-regulated in the brain tissues from AD patients and 5XFAD mice. The addition of miR-188-5p rescued the reduction in dendritic spine density in the primary hippocampal neurons treated with oligomeric Aβ1-42 and cultured from 5XFAD mice. The reduction in the frequency of mEPSCs was also restored by addition of miR-188-5p. The impairments in basal fEPSPs and cognition observed in 7-month-old 5XFAD mice were ameliorated via the viral-mediated expression of miR-188-5p in the hippocampus. Furthermore, we found that miR-188 expression is CREB-dependent. Taken together, our results suggest that dysregulation of miR-188-5p expression contributes to the pathogenesis of AD by inducing synaptic dysfunction and cognitive deficits associated with Aβ-mediated pathophysiology in the disease.
Collapse
Affiliation(s)
- Kihwan Lee
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Hyunju Kim
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Kyongman An
- Department of Life Science, POSTECH, Pohang, Gyeongbuk, 790-784, Republic of Korea
| | - Oh-Bin Kwon
- Department of Life Science, POSTECH, Pohang, Gyeongbuk, 790-784, Republic of Korea
| | - Sungjun Park
- Department of Life Science, POSTECH, Pohang, Gyeongbuk, 790-784, Republic of Korea
| | - Jin Hee Cha
- Department of Physiology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Myoung-Hwan Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Yoontae Lee
- Department of Life Science, POSTECH, Pohang, Gyeongbuk, 790-784, Republic of Korea
| | - Joung-Hun Kim
- Department of Life Science, POSTECH, Pohang, Gyeongbuk, 790-784, Republic of Korea
| | - Kwangwook Cho
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology (LINE), Faculty of Medicine and Dentistry, University of Bristol, Whitson Street, Bristol BS1 3NY, UK.,Centre for Synaptic Plasticity, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | - Hye-Sun Kim
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea.,Seoul National University Bundang Hospital, College of Medicine, Seoul National University, Seongnam, Gyeonggi, 463-707, Republic of Korea
| |
Collapse
|
35
|
Xi YD, Zhang DD, Ding J, Yu HL, Yuan LH, Ma WW, Han J, Xiao R. Genistein Inhibits Aβ25-35-Induced Synaptic Toxicity and Regulates CaMKII/CREB Pathway in SH-SY5Y Cells. Cell Mol Neurobiol 2016; 36:1151-9. [PMID: 26658733 PMCID: PMC11482296 DOI: 10.1007/s10571-015-0311-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/24/2015] [Indexed: 02/07/2023]
Abstract
Genistein (Gen), as a functional food in human diet, has shown many beneficial effects on neurodegenerative diseases such as Alzheimer's disease (AD). But the neuroprotective mechanism of Gen is not clear. Because synaptic failure is considered as the earliest phase in the pathogenesis of AD, we try to validate our hypothesis that synapse may be one target of Gen on protecting neurons. In this study, SH-SY5Y cells were pre-incubated with or without Gen for 2 h followed by the incubation with Aβ25-35 (25 μmol/L) for another 24 h. Flow cytometry, Western Blots, and RT-PCR analysis were used to test the synaptic factors. The data showed that Gen pre-treatment could reverse the Aβ25-35-induced down-regulation of synaptophysin and postsynaptic marker postsynaptic density-95. In addition, the down-regulation of NR1 and NR2B induced by Aβ25-35 which are subunits of N-methyl-D-aspartate receptor also could be antagonized by pre-treatment of Gen. Moreover, the factors of CaMKII/CREB signaling pathway were detected. The results showed that mRNA and protein expressions of (Ca(2+))/calmodulin(CaM), CaMKII/pCaMKII, and CREB/pCREB were significantly down-regulated by Aβ25-35, but they were all restored by the pre-treatment of Gen. Furthermore, Gen also maintained the intracellular Ca(2+) concentration which was disturbed by Aβ25-35. In conclusion, these results suggested that Gen could protect synaptic dysfunction induced by Aβ, and the mechanism might be associated with the regulation of synaptic markers and Ca(2+) level through activating CaM/CaMK/CREB signaling pathway.
Collapse
Affiliation(s)
- Yuan-Di Xi
- School of Public Health, Capital Medical University and Beijing Key Laboratory of Environmental Toxicology, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China
| | - Dan-Di Zhang
- School of Public Health, Capital Medical University and Beijing Key Laboratory of Environmental Toxicology, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China
| | - Juan Ding
- Ningxia Key Laboratory of Cerebrocranial Diseases, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Huan-Ling Yu
- School of Public Health, Capital Medical University and Beijing Key Laboratory of Environmental Toxicology, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China
| | - Lin-Hong Yuan
- School of Public Health, Capital Medical University and Beijing Key Laboratory of Environmental Toxicology, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China
| | - Wei-Wei Ma
- School of Public Health, Capital Medical University and Beijing Key Laboratory of Environmental Toxicology, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China
| | - Jing Han
- School of Public Health, Capital Medical University and Beijing Key Laboratory of Environmental Toxicology, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China
| | - Rong Xiao
- School of Public Health, Capital Medical University and Beijing Key Laboratory of Environmental Toxicology, No. 10 Xitoutiao, You An Men Wai, Beijing, 100069, China.
| |
Collapse
|
36
|
Gu M, Viles JH. Methionine oxidation reduces lag-times for amyloid-β(1-40) fiber formation but generates highly fragmented fibers. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1260-1269. [PMID: 27108954 DOI: 10.1016/j.bbapap.2016.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/19/2016] [Indexed: 01/06/2023]
Abstract
Oxidative stress and the formation of amyloid plaques containing amyloid-β (Aβ) peptides are two key hallmarks of Alzheimer's disease. A proportion of methionine (Met) at position 35 within Aβ is oxidized to methionine sulphoxide (Met(OX)) within the Alzheimer's plaques. These oxidative processes may be the key to understanding the early stages of Alzheimer's disease. In vitro oxidation of Aβ, by the physiological oxidant H2O2, was monitored using (1)H NMR and mass spectrometry. Here we investigate the effect of Aβ methionine oxidation on fiber formation kinetics and morphology using the amyloid specific fluorescence dye Thioflavin T (ThT) and Transmission Electron Microscopy (TEM). Methionine oxidation reduces the total amount of fibers generated for both dominant forms of Aβ, however there are marked differences in the effect of Met(OX) between Aβ(1-40) and Aβ(1-42). Surprisingly the presence of Met(OX) reduces lag-times for Aβ(1-40) fiber formation but extends lag-times for Aβ(1-42). TEM indicates a change in fiber morphology with a pronounced reduction in fiber length for both methionine oxidized Aβ(1-40) and Aβ(1-42). In contrast, the morphology of preformed amyloid fibers is largely unaffected by the presence of H2O2. Our studies suggest that methionine oxidation promotes highly fragmented fiber assemblies of Aβ. Oxidative stress associated with Alzheimer's disease can cause oxidation of methionine within Aβ and this in turn will influence the complex assembly of Aβ monomer into amyloid fibers, which is likely to impact Aβ toxicity.
Collapse
Affiliation(s)
- Miao Gu
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, United Kingdom
| | - John H Viles
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, United Kingdom
| |
Collapse
|
37
|
Das AK, Pandit R, Maiti S. Effect of amyloids on the vesicular machinery: implications for somatic neurotransmission. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0187. [PMID: 26009766 DOI: 10.1098/rstb.2014.0187] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Certain neurodegenerative diseases are thought to be initiated by the aggregation of amyloidogenic proteins. However, the mechanism underlying toxicity remains obscure. Most of the suggested mechanisms are generic in nature and do not directly explain the neuron-type specific lesions observed in many of these diseases. Some recent reports suggest that the toxic aggregates impair the synaptic vesicular machinery. This may lead to an understanding of the neuron-type specificity observed in these diseases. A disruption of the vesicular machinery can also be deleterious for extra-synaptic, especially somatic, neurotransmission (common in serotonergic and dopaminergic systems which are specifically affected in Alzheimer's disease (AD) and Parkinson's disease (PD), respectively), though this relationship has remained unexplored. In this review, we discuss amyloid-induced damage to the neurotransmitter vesicular machinery, with an eye on the possible implications for somatic exocytosis. We argue that the larger size of the system, and the availability of multi-photon microscopy techniques for directly visualizing monoamines, make the somatic exocytosis machinery a more tractable model for understanding the effect of amyloids on all types of vesicular neurotransmission. Indeed, exploring this neglected connection may not just be important, it may be a more fruitful route for understanding AD and PD.
Collapse
Affiliation(s)
- Anand Kant Das
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, Maharashtra 400005, India
| | - Rucha Pandit
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, Maharashtra 400005, India
| | - Sudipta Maiti
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, Maharashtra 400005, India
| |
Collapse
|
38
|
Fernández-Fernández D, Dorner-Ciossek C, Kroker KS, Rosenbrock H. Age-related synaptic dysfunction in Tg2576 mice starts as a failure in early long-term potentiation which develops into a full abolishment of late long-term potentiation. J Neurosci Res 2015; 94:266-81. [PMID: 26629777 DOI: 10.1002/jnr.23701] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/30/2015] [Accepted: 11/17/2015] [Indexed: 11/12/2022]
Abstract
Tg2576 mice are widely used to study amyloid-dependent synaptic dysfunction related to Alzheimer's disease. However, conflicting data have been reported for these mice with regard to basal transmission as well as the in vitro correlate of memory, long-term potentiation (LTP). Some studies show clear impairments, whereas others report no deficiency. The present study uses hippocampal slices from 3-, 10-, and 15-month-old wild-type (WT) and Tg2576 mice to evaluate synaptic function in each group, including experiments to investigate basal synaptic transmission, short- and long-term plasticity by inducing paired-pulse facilitation, and both early and late LTP. We show that synaptic function remains intact in hippocampal slices from Tg2576 mice at 3 months of age. However, both early and late LTP decline progressively during aging in these mice. This deterioration of synaptic plasticity starts affecting early LTP, ultimately leading to the abolishment of both forms of LTP in 15-month-old animals. In comparison, WT littermates display normal synaptic parameters during aging. Additional pharmacological investigation into the involvement of NMDA receptors and L-type voltage-gated calcium channels in LTP suggests a distinct mechanism of induction among age groups, demonstrating that both early and late LTP are differentially affected by these channels in Tg2576 mice during aging.
Collapse
Affiliation(s)
- Diego Fernández-Fernández
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Cornelia Dorner-Ciossek
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Katja S Kroker
- Deptartment of Drug Discovery Support, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Holger Rosenbrock
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| |
Collapse
|
39
|
Herpes Simplex Virus type-1 infection induces synaptic dysfunction in cultured cortical neurons via GSK-3 activation and intraneuronal amyloid-β protein accumulation. Sci Rep 2015; 5:15444. [PMID: 26487282 PMCID: PMC4614347 DOI: 10.1038/srep15444] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/15/2015] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence suggests that recurrent Herpes Simplex Virus type 1 (HSV-1) infection spreading to the CNS is a risk factor for Alzheimer’s Disease (AD) but the underlying mechanisms have not been fully elucidated yet. Here we demonstrate that in cultured mouse cortical neurons HSV-1 induced Ca2+-dependent activation of glycogen synthase kinase (GSK)-3. This event was critical for the HSV-1-dependent phosphorylation of amyloid precursor protein (APP) at Thr668 and the following intraneuronal accumulation of amyloid-β protein (Aβ). HSV-1-infected neurons also exhibited: i) significantly reduced expression of the presynaptic proteins synapsin-1 and synaptophysin; ii) depressed synaptic transmission. These effects depended on GSK-3 activation and intraneuronal accumulation of Aβ. In fact, either the selective GSK-3 inhibitor, SB216763, or a specific antibody recognizing Aβ (4G8) significantly counteracted the effects induced by HSV-1 at the synaptic level. Moreover, in neurons derived from APP KO mice and infected with HSV-1 Aβ accumulation was not found and synaptic protein expression was only slightly reduced when compared to wild-type infected neurons. These data further support our contention that HSV-1 infections spreading to the CNS may contribute to AD phenotype.
Collapse
|
40
|
Tamagnini F, Novelia J, Kerrigan TL, Brown JT, Tsaneva-Atanasova K, Randall AD. Altered intrinsic excitability of hippocampal CA1 pyramidal neurons in aged PDAPP mice. Front Cell Neurosci 2015; 9:372. [PMID: 26528126 PMCID: PMC4604241 DOI: 10.3389/fncel.2015.00372] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/07/2015] [Indexed: 12/28/2022] Open
Abstract
Amyloidopathy involves the accumulation of insoluble amyloid β (Aβ) species in the brain's parenchyma and is a key histopathological hallmark of Alzheimer's disease (AD). Work on transgenic mice that overexpress Aβ suggests that elevated Aβ levels in the brain are associated with aberrant epileptiform activity and increased intrinsic excitability (IE) of CA1 hippocampal neurons. In this study we examined if similar changes could be observed in hippocampal CA1 pyramidal neurons from aged PDAPP mice (20-23 month old, Indiana mutation: V717F on APP gene) compared to their age-matched wild-type littermate controls. Whole-cell current clamp recordings revealed that sub-threshold intrinsic properties, such as input resistance, resting membrane potential and hyperpolarization activated "sag" were unaffected, but capacitance was significantly decreased in the transgenic animals. No differences between genotypes were observed in the overall number of action potentials (AP) elicited by 500 ms supra-threshold current stimuli. PDAPP neurons, however, exhibited higher instantaneous firing frequencies after accommodation in response to high intensity current injections. The AP waveform was narrower and shorter in amplitude in PDAPP mice: these changes, according to our in silico model of a CA1/3 pyramidal neuron, depended on the respective increase and reduction of K(+) and Na(+) voltage-gated channels maximal conductances. Finally, the after-hyperpolarization, seen after the first AP evoked by a +300 pA current injection and after 50 Hz AP bursts, was more pronounced in PDAPP mice. These data show that Aβ-overexpression in aged mice altered the capacitance, the neuronal firing and the AP waveform of CA1 pyramidal neurons. Some of these findings are consistent with previous work on younger PDAPP; they also show important differences that can be potentially ascribed to the interaction between amyloidopathy and ageing. Such a change of IE properties over time underlies that the increased incidence of seizure observed in AD patients might rely on different mechanistic pathways during progression of the disease.
Collapse
Affiliation(s)
- Francesco Tamagnini
- Medical School, University of Exeter Exeter, UK ; School of Physiology and Pharmacology, University of Bristol Bristol, UK
| | - Janet Novelia
- Department of Mathematics, College of Engineering, Mathematics and Physical Sciences, University of Exeter Exeter, UK
| | - Talitha L Kerrigan
- Medical School, University of Exeter Exeter, UK ; School of Physiology and Pharmacology, University of Bristol Bristol, UK
| | - Jon T Brown
- Medical School, University of Exeter Exeter, UK ; School of Physiology and Pharmacology, University of Bristol Bristol, UK
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics, College of Engineering, Mathematics and Physical Sciences, University of Exeter Exeter, UK
| | - Andrew D Randall
- Medical School, University of Exeter Exeter, UK ; School of Physiology and Pharmacology, University of Bristol Bristol, UK
| |
Collapse
|
41
|
Cai ZX, Guo HS, Wang C, Wei M, Cheng C, Yang ZF, Chen YW, Le WD, Li S. Double-Edged Roles of Nitric Oxide Signaling on APP Processing and Amyloid-β Production In Vitro: Preliminary Evidence from Sodium Nitroprusside. Neurotox Res 2015; 29:21-34. [PMID: 26429731 DOI: 10.1007/s12640-015-9564-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 09/02/2015] [Accepted: 09/22/2015] [Indexed: 11/27/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is thought to be caused in part by the age-related accumulation of amyloid-β (Aβ) in the brain. Recent findings have revealed that nitric oxide (NO) modulates the processing of amyloid-β precursor protein (APP) and alters Aβ production; however, the previously presented data are contradictory and the underlying molecular mechanisms are still incomplete. Here, using human SH-SY5Y neuroblastoma cells stably transfected with wild-type APPwt695, we found that NO, derived from NO donor sodium nitroprusside (SNP), bi-directionally modulates APP processing in vitro. The data from ELISA and Western blot (WB) tests indicated that SNP at lower concentrations (0.01 and 0.1 μM) inhibits BACE1 expression, thus consequently suppresses APP β-cleavage and decreases Aβ production. In contrast, SNP at higher concentrations (10 and 20 μM) biases the APP processing toward the amyloidogenic pathway as evidenced by an increased BACE1 but a decreased ADAM10 expression, together with an elevated Aβ secretion. This bi-directional modulating activity of SNP on APP processing was completely blocked by specific NO scavenger c-PTIO, indicating NO-dependent mechanisms. Moreover, the anti-amyloidogenic activity of SNP is sGC/cGMP/PKG-dependent as evidenced by its reversal by sGC/PKG inhibitions, whereas the amyloidogenic activity of SNP is peroxynitrite-related and can be reversed by peroxynitrite scavenger uric acid. In summary, these present findings predict a double-edged role of NO in APP processing in vitro. Low (physiological) levels of NO inhibit the amyloidogenic processing of APP, whereas extra-high (pathological) concentrations of NO favor the amyloidogenic pathway of APP processing. This preliminary study may provide further evidence to clarify the molecular roles of NO and NO-related signaling in AD and supply potential molecular targets for AD treatment.
Collapse
Affiliation(s)
- Zheng-Xu Cai
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Hui-Shu Guo
- Laboratory Center, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Che Wang
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China.,Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University, Dalian, 116029, China
| | - Min Wei
- Center for Translational Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Cheng Cheng
- Center for Translational Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Zhao-Fei Yang
- Center for Translational Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Yin-Wang Chen
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Wei-Dong Le
- Center for Translational Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China.
| | - Song Li
- Center for Translational Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
42
|
Yang EJ, Ahn S, Ryu J, Choi MS, Choi S, Chong YH, Hyun JW, Chang MJ, Kim HS. Phloroglucinol Attenuates the Cognitive Deficits of the 5XFAD Mouse Model of Alzheimer's Disease. PLoS One 2015; 10:e0135686. [PMID: 26284625 PMCID: PMC4540482 DOI: 10.1371/journal.pone.0135686] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 07/26/2015] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia among the elderly. Neuritic plaques whose primary component is amyloid beta peptide (Aβ) and neurofibrillary tangles which are composed of hyperphosphorylated tau, are known to be the neuropathological hallmarks of AD. In addition, impaired synaptic plasticity in neuronal networks is thought to be important mechanism underlying for the cognitive deficits observed in AD. Although various causative factors, including excitotoxicity, mitochondrial dysregulation and oxidative damage caused by Aβ, are involved in early onset of AD, fundamental therapeutics that can modify the progression of this disease are not currently available. In the present study, we investigated whether phloroglucinol (1, 3, 5-trihydroxybenzene), a component of phlorotannins, which are plentiful in Ecklonia cava, a marine brown alga species, displays therapeutic activities in AD. We found that phloroglucinol attenuates the increase in reactive oxygen species (ROS) accumulation induced by oligomeric Aβ1-42 (Aβ1-42) treatment in HT-22, hippocampal cell line. In addition, phloroglucinol was shown to ameliorate the reduction in dendritic spine density induced by Aβ1-42 treatment in rat primary hippocampal neuron cultures. We also found that the administration of phloroglucinol to the hippocampal region attenuated the impairments in cognitive dysfunction observed in 22-week-old 5XFAD (Tg6799) mice, which are used as an AD animal model. These results indicate that phloroglucinol displays therapeutic potential for AD by reducing the cellular ROS levels.
Collapse
Affiliation(s)
- Eun-Jeong Yang
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sangzin Ahn
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Junghwa Ryu
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Moon-Seok Choi
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Shinkyu Choi
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Young Hae Chong
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Jin-Won Hyun
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Moon-Jeong Chang
- Department of Foods and Nutrition, College of Natural Science, Kookmin University, Seoul, Republic of Korea
| | - Hye-Sun Kim
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Seoul National University College of Medicine, Bundang Hospital, Bundang-Gu, Sungnam, Republic of Korea
- Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Friedemann M, Helk E, Tiiman A, Zovo K, Palumaa P, Tõugu V. Effect of methionine-35 oxidation on the aggregation of amyloid-β peptide. Biochem Biophys Rep 2015; 3:94-99. [PMID: 29124171 PMCID: PMC5668694 DOI: 10.1016/j.bbrep.2015.07.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 07/22/2015] [Accepted: 07/28/2015] [Indexed: 01/12/2023] Open
Abstract
Aggregation of Aβ peptides into amyloid plaques is considered to trigger the Alzheimer’s disease (AD), however the mechanism behind the AD onset has remained elusive. It is assumed that the insoluble Aβ aggregates enhance oxidative stress (OS) by generating free radicals with the assistance of bound copper ions. The aim of our study was to establish the role of Met35 residue in the oxidation and peptide aggregation processes. Met35 can be readily oxidized by H2O2. The fibrillization of Aβ with Met35 oxidized to sulfoxide was three times slower compared to that of the regular peptide. The fibrils of regular and oxidized peptides looked similar under transmission electron microscopy. The relatively small inhibitory effect of methionine oxidation on the fibrillization suggests that the possible variation in the Met oxidation state should not affect the in vivo plaque formation. The peptide oxidation pattern was more complex when copper ions were present: addition of one oxygen atom was still the fastest process, however, it was accompanied by multiple unspecific modifications of peptide residues. Addition of copper ions to the Aβ with oxidized Met35 in the presence of H2O2, resulted a similar pattern of nonspecific modifications, suggesting that the one-electron oxidation processes in the peptide molecule do not depend on the oxidation state of Met35 residue. Thus, it can be concluded that Met35 residue is not a part of the radical generating mechanism of Aβ–Cu(II) complex. Aβ peptides with oxidized Met35 residue fibrillize three times slower than the reduced peptide. Met35 is the only residue in Aβ peptide that is oxidized by H2O2 in the absent of copper ions. In the presence of copper ions as catalyst multiple unspecific oxidative processes occur in Aβ. Previous oxidation of Met35 does not affect the unspecific oxidation in the presence of copper ions.
Collapse
Key Words
- AD, Alzheimer's disease
- Alzheimer's disease
- Aβ, Alzheimer's amyloid peptide
- Copper(II)ion
- HFIP, 1,1,1,3,3,3-hexafluoro-2-propanol
- Methionine oxidation
- OS, oxidative stress
- ROS, reactive oxygen species
- ThT, Thioflavin T
- β-amyloid
Collapse
Affiliation(s)
- Merlin Friedemann
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Eneken Helk
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Ann Tiiman
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Kairit Zovo
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Peep Palumaa
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Vello Tõugu
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| |
Collapse
|
44
|
Chegaev K, Federico A, Marini E, Rolando B, Fruttero R, Morbin M, Rossi G, Fugnanesi V, Bastone A, Salmona M, Badiola NB, Gasparini L, Cocco S, Ripoli C, Grassi C, Gasco A. NO-donor thiacarbocyanines as multifunctional agents for Alzheimer's disease. Bioorg Med Chem 2015; 23:4688-4698. [PMID: 26078011 DOI: 10.1016/j.bmc.2015.05.050] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/26/2015] [Accepted: 05/29/2015] [Indexed: 12/31/2022]
Abstract
Some symmetrical and unsymmetrical thiacarbocyanines bearing NO-donor nitrooxy and furoxan moieties were synthesized and studied as candidate anti-Alzheimer's drugs. All products activated soluble guanylate cyclase (sGC) in a dose-dependent manner, depending on the presence in their structures of NO-donor groups. None displayed toxicity when tested at concentrations below 10 μM on human brain microvascular endothelial cells (hCMEC/D3). Some products were capable of inhibiting amyloid β-protein (Aβ) aggregation, with a potency in the low μM concentration range, and of inhibiting aggregation of human recombinant tau protein in amyloid fibrils when incubated with the protein at 1 μM concentration. Nitrooxy derivative 21 and furoxan derivative 22 were selected to investigate synaptic plasticity. Both products, tested at 2 μM concentration, counteracted the inhibition of long-term potentiation (LTP) induced by Aβ42 in hippocampal brain slices.
Collapse
Affiliation(s)
- Konstantin Chegaev
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
| | - Antonella Federico
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
| | - Elisabetta Marini
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
| | - Barbara Rolando
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
| | - Roberta Fruttero
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy.
| | - Michela Morbin
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Giacomina Rossi
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Valeria Fugnanesi
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Antonio Bastone
- Department of Molecular Biochemistry and Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche 'Mario Negri', Milano, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche 'Mario Negri', Milano, Italy
| | - Nahuai B Badiola
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| | - Laura Gasparini
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| | - Sara Cocco
- Institute of Human Physiology, UniversitàCattolica, Roma, Italy
| | - Cristian Ripoli
- Institute of Human Physiology, UniversitàCattolica, Roma, Italy
| | - Claudio Grassi
- Institute of Human Physiology, UniversitàCattolica, Roma, Italy
| | - Alberto Gasco
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, via Pietro Giuria 9, 10125 Torino, Italy
| |
Collapse
|
45
|
Scala F, Fusco S, Ripoli C, Piacentini R, Li Puma DD, Spinelli M, Laezza F, Grassi C, D'Ascenzo M. Intraneuronal Aβ accumulation induces hippocampal neuron hyperexcitability through A-type K(+) current inhibition mediated by activation of caspases and GSK-3. Neurobiol Aging 2015; 36:886-900. [PMID: 25541422 PMCID: PMC4801354 DOI: 10.1016/j.neurobiolaging.2014.10.034] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 10/14/2014] [Accepted: 10/24/2014] [Indexed: 11/20/2022]
Abstract
Amyloid β-protein (Aβ) pathologies have been linked to dysfunction of excitability in neurons of the hippocampal circuit, but the molecular mechanisms underlying this process are still poorly understood. Here, we applied whole-cell patch-clamp electrophysiology to primary hippocampal neurons and show that intracellular Aβ42 delivery leads to increased spike discharge and action potential broadening through downregulation of A-type K(+) currents. Pharmacologic studies showed that caspases and glycogen synthase kinase 3 (GSK-3) activation are required for these Aβ42-induced effects. Extracellular perfusion and subsequent internalization of Aβ42 increase spike discharge and promote GSK-3-dependent phosphorylation of the Kv4.2 α-subunit, a molecular determinant of A-type K(+) currents, at Ser-616. In acute hippocampal slices derived from an adult triple-transgenic Alzheimer's mouse model, characterized by endogenous intracellular accumulation of Aβ42, CA1 pyramidal neurons exhibit hyperexcitability accompanied by increased phosphorylation of Kv4.2 at Ser-616. Collectively, these data suggest that intraneuronal Aβ42 accumulation leads to an intracellular cascade culminating into caspases activation and GSK-3-dependent phosphorylation of Kv4.2 channels. These findings provide new insights into the toxic mechanisms triggered by intracellular Aβ42 and offer potentially new therapeutic targets for Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Federico Scala
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | - Salvatore Fusco
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | - Cristian Ripoli
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | - Roberto Piacentini
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | | | - Matteo Spinelli
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy.
| | - Marcello D'Ascenzo
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy.
| |
Collapse
|
46
|
Minniti AN, Arrazola MS, Bravo-Zehnder M, Ramos F, Inestrosa NC, Aldunate R. The protein oxidation repair enzyme methionine sulfoxide reductase a modulates Aβ aggregation and toxicity in vivo. Antioxid Redox Signal 2015; 22:48-62. [PMID: 24988428 PMCID: PMC4270145 DOI: 10.1089/ars.2013.5803] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS To examine the role of the enzyme methionine sulfoxide reductase A-1 (MSRA-1) in amyloid-β peptide (Aβ)-peptide aggregation and toxicity in vivo, using a Caenorhabditis elegans model of the human amyloidogenic disease inclusion body myositis. RESULTS MSRA-1 specifically reduces oxidized methionines in proteins. Therefore, a deletion of the msra-1 gene was introduced into transgenic C. elegans worms that express the Aβ-peptide in muscle cells to prevent the reduction of oxidized methionines in proteins. In a constitutive transgenic Aβ strain that lacks MSRA-1, the number of amyloid aggregates decreases while the number of oligomeric Aβ species increases. These results correlate with enhanced synaptic dysfunction and mislocalization of the nicotinic acetylcholine receptor ACR-16 at the neuromuscular junction (NMJ). INNOVATION This approach aims at modulating the oxidation of Aβ in vivo indirectly by dismantling the methionine sulfoxide repair system. The evidence presented here shows that the absence of MSRA-1 influences Aβ aggregation and aggravates locomotor behavior and NMJ dysfunction. The results suggest that therapies which boost the activity of the Msr system could have a beneficial effect in managing amyloidogenic pathologies. CONCLUSION The absence of MSRA-1 modulates Aβ-peptide aggregation and increments its deleterious effects in vivo.
Collapse
Affiliation(s)
- Alicia N Minniti
- 1 Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago, Chile
| | | | | | | | | | | |
Collapse
|
47
|
Nenov MN, Tempia F, Denner L, Dineley KT, Laezza F. Impaired firing properties of dentate granule neurons in an Alzheimer's disease animal model are rescued by PPARγ agonism. J Neurophysiol 2014; 113:1712-26. [PMID: 25540218 PMCID: PMC4359997 DOI: 10.1152/jn.00419.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Early cognitive impairment in Alzheimer's disease (AD) correlates with medial temporal lobe dysfunction, including two areas essential for memory formation: the entorhinal cortex and dentate gyrus (DG). In the Tg2576 animal model for AD amyloidosis, activation of the peroxisome proliferator-activated receptor-gamma (PPARγ) with rosiglitazone (RSG) ameliorates hippocampus-dependent cognitive impairment and restores aberrant synaptic activity at the entorhinal cortex to DG granule neuron inputs. It is unknown, however, whether intrinsic firing properties of DG granule neurons in these animals are affected by amyloid-β pathology and if they are sensitive to RSG treatment. Here, we report that granule neurons from 9-mo-old wild-type and Tg2576 animals can be segregated into two cell types with distinct firing properties and input resistance that correlate with less mature type I and more mature type II neurons. The DG type I cell population was greater than type II in wild-type littermates. In the Tg2576 animals, the type I and type II cell populations were nearly equal but could be restored to wild-type levels through cognitive enhancement with RSG. Furthermore, Tg2576 cell firing frequency and spike after depolarization were decreased in type I and increased in type II cells, both of which could also be restored to wild-type levels upon RSG treatment. That these parameters were restored by PPARγ activation emphasizes the therapeutic value of RSG against early AD cognitive impairment.
Collapse
Affiliation(s)
- Miroslav N Nenov
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas
| | - Filippo Tempia
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas
| | - Larry Denner
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas; Center for Addiction Research, The University of Texas Medical Branch, Galveston, Texas; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, Texas; and
| | - Kelly T Dineley
- Department of Neurology, The University of Texas Medical Branch, Galveston, Texas; Center for Addiction Research, The University of Texas Medical Branch, Galveston, Texas; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, Texas; and
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas; Center for Addiction Research, The University of Texas Medical Branch, Galveston, Texas; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, Texas; and Center for Biomedical Engineering, The University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
48
|
Intracellular accumulation of amyloid-β (Aβ) protein plays a major role in Aβ-induced alterations of glutamatergic synaptic transmission and plasticity. J Neurosci 2014; 34:12893-903. [PMID: 25232124 DOI: 10.1523/jneurosci.1201-14.2014] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Intracellular accumulation of amyloid-β (Aβ) protein has been proposed as an early event in AD pathogenesis. In patients with mild cognitive impairment, intraneuronal Aβ immunoreactivity was found especially in brain regions critically involved in the cognitive deficits of AD. Although a large body of evidence demonstrates that Aβ42 accumulates intraneuronally ((in)Aβ), the action and the role of Aβ42 buildup on synaptic function have been poorly investigated. Here, we demonstrate that basal synaptic transmission and LTP were markedly depressed following Aβ42 injection into the neuron through the patch pipette. Control experiments performed with the reverse peptide (Aβ42-1) allowed us to exclude that the effects of (in)Aβ depended on changes in oncotic pressure. To further investigate (in)Aβ synaptotoxicity we used an Aβ variant harboring oxidized methionine in position 35 that does not cross the neuronal plasma membrane and is not uploaded from the extracellular space. This Aβ42 variant had no effects on synaptic transmission and plasticity when applied extracellularly, but induced synaptic depression and LTP inhibition after patch-pipette dialysis. Finally, the injection of an antibody raised against human Aβ42 (6E10) in CA1 pyramidal neurons of mouse hippocampal brain slices and autaptic microcultures did not, per se, significantly affect LTP and basal synaptic transmission, but it protected against the toxic effects of extracellular Aβ42. Collectively, these findings suggest that Aβ42-induced impairment of glutamatergic synaptic function depends on its internalization and intracellular accumulation thus paving the way to a systemic proteomic analysis of intracellular targets/partners of Aβ42.
Collapse
|
49
|
Piacentini R, De Chiara G, Li Puma DD, Ripoli C, Marcocci ME, Garaci E, Palamara AT, Grassi C. HSV-1 and Alzheimer's disease: more than a hypothesis. Front Pharmacol 2014; 5:97. [PMID: 24847267 PMCID: PMC4019841 DOI: 10.3389/fphar.2014.00097] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/16/2014] [Indexed: 12/22/2022] Open
Abstract
Among the multiple factors concurring to Alzheimer’s disease (AD) pathogenesis, greater attention should be devoted to the role played by infectious agents. Growing epidemiological and experimental evidence suggests that recurrent herpes simplex virus type-1 (HSV-1) infection is a risk factor for AD although the underlying molecular and functional mechanisms have not been fully elucidated yet. Here, we review literature suggesting the involvement of HSV-1 infection in AD also briefly mentioning possible pharmacological implications of these findings.
Collapse
Affiliation(s)
- Roberto Piacentini
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| | - Giovanna De Chiara
- Institute of Translational Pharmacology, National Research Council Rome, Italy
| | - Domenica D Li Puma
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| | - Cristian Ripoli
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| | - Maria E Marcocci
- Department of Public Health and Infectious Diseases, Sapienza University of Rome Rome, Italy
| | - Enrico Garaci
- San Raffaele Pisana Scientific Institute for Research, Hospitalization and Health Care, Telematic University Rome, Italy
| | - Anna T Palamara
- Department of Public Health and Infectious Diseases, Institute Pasteur Cenci Bolognetti Foundation, Sapienza University of Rome Rome, Italy ; San Raffaele Pisana Scientific Institute for Research, Hospitalization and Health Care Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| |
Collapse
|
50
|
Cognitive enhancing treatment with a PPARγ agonist normalizes dentate granule cell presynaptic function in Tg2576 APP mice. J Neurosci 2014; 34:1028-36. [PMID: 24431460 DOI: 10.1523/jneurosci.3413-13.2014] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hippocampal network hyperexcitability is considered an early indicator of Alzheimer's disease (AD) memory impairment. Some AD mouse models exhibit similar network phenotypes. In this study we focused on dentate gyrus (DG) granule cell spontaneous and evoked properties in 9-month-old Tg2576 mice that model AD amyloidosis and cognitive deficits. Using whole-cell patch-clamp recordings, we found that Tg2576 DG granule cells exhibited spontaneous EPSCs that were higher in frequency but not amplitude compared with wild-type mice, suggesting hyperactivity of DG granule cells via a presynaptic mechanism. Further support of a presynaptic mechanism was revealed by increased I-O relationships and probability of release in Tg2576 DG granule cells. Since we and others have shown that activation of the peroxisome proliferator-activated receptor gamma (PPARγ) axis improves hippocampal cognition in mouse models for AD as well as benefitting memory performance in some humans with early AD, we investigated how PPARγ agonism affected synaptic activity in Tg2576 DG. We found that PPARγ agonism normalized the I-O relationship of evoked EPSCs, frequency of spontaneous EPSCs, and probability of release that, in turn, correlated with selective expression of DG proteins essential for presynaptic SNARE function that are altered in patients with AD. These findings provide evidence that DG principal cells may contribute to early AD hippocampal network hyperexcitability via a presynaptic mechanism, and that hippocampal cognitive enhancement via PPARγ activation occurs through regulation of presynaptic vesicular proteins critical for proper glutamatergic neurotransmitter release, synaptic transmission, and short-term plasticity.
Collapse
|