1
|
Milham LT, Morris GP, Konen LM, Rentsch P, Avgan N, Vissel B. Quantification of AMPA receptor subunits and RNA editing-related proteins in the J20 mouse model of Alzheimer's disease by capillary western blotting. Front Mol Neurosci 2024; 16:1338065. [PMID: 38299128 PMCID: PMC10828003 DOI: 10.3389/fnmol.2023.1338065] [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: 11/14/2023] [Accepted: 12/29/2023] [Indexed: 02/02/2024] Open
Abstract
Introduction Accurate modelling of molecular changes in Alzheimer's disease (AD) dementia is crucial for understanding the mechanisms driving neuronal pathology and for developing treatments. Synaptic dysfunction has long been implicated as a mechanism underpinning memory dysfunction in AD and may result in part from changes in adenosine deaminase acting on RNA (ADAR) mediated RNA editing of the GluA2 subunit of AMPA receptors and changes in AMPA receptor function at the post synaptic cleft. However, few studies have investigated changes in proteins which influence RNA editing and notably, AD studies that focus on studying changes in protein expression, rather than changes in mRNA, often use traditional western blotting. Methods Here, we demonstrate the value of automated capillary western blotting to investigate the protein expression of AMPA receptor subunits (GluA1-4), the ADAR RNA editing proteins (ADAR1-3), and proteins known to regulate RNA editing (PIN1, WWP2, FXR1P, and CREB1), in the J20 AD mouse model. We describe extensive optimisation and validation of the automated capillary western blotting method, demonstrating the use of total protein to normalise protein load, in addition to characterising the optimal protein/antibody concentrations to ensure accurate protein quantification. Following this, we assessed changes in proteins of interest in the hippocampus of 44-week-old J20 AD mice. Results We observed an increase in the expression of ADAR1 p110 and GluA3 and a decrease in ADAR2 in the hippocampus of 44-week-old J20 mice. These changes signify a shift in the balance of proteins that play a critical role at the synapse. Regression analysis revealed unique J20-specific correlations between changes in AMPA receptor subunits, ADAR enzymes, and proteins that regulate ADAR stability in J20 mice, highlighting potential mechanisms mediating RNA-editing changes found in AD. Discussion Our findings in J20 mice generally reflect changes seen in the human AD brain. This study underlines the importance of novel techniques, like automated capillary western blotting, to assess protein expression in AD. It also provides further evidence to support the hypothesis that a dysregulation in RNA editing-related proteins may play a role in the initiation and/or progression of AD.
Collapse
Affiliation(s)
- Luke T. Milham
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Gary P. Morris
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Lyndsey M. Konen
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Peggy Rentsch
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Nesli Avgan
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Bryce Vissel
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
2
|
Rathod SS, Agrawal YO. Phytocannabinoids as Potential Multitargeting Neuroprotectants in Alzheimer's Disease. Curr Drug Res Rev 2024; 16:94-110. [PMID: 37132109 DOI: 10.2174/2589977515666230502104021] [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: 10/20/2022] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 05/04/2023]
Abstract
The Endocannabinoid System (ECS) is a well-studied system that influences a variety of physiological activities. It is evident that the ECS plays a significant role in metabolic activities and also has some neuroprotective properties. In this review, we emphasize several plant-derived cannabinoids such as β-caryophyllene (BCP), Cannabichromene (CBC), Cannabigerol (CBG), Cannabidiol (CBD), and Cannabinol (CBN), which are known to have distinctive modulation abilities of ECS. In Alzheimer's disease (AD), the activation of ECS may provide neuroprotection by modulating certain neuronal circuitry pathways through complex molecular cascades. The present article also discusses the implications of cannabinoid receptors (CB1 and CB2) as well as cannabinoid enzymes (FAAH and MAGL) modulators in AD. Specifically, CBR1 or CB2R modulations result in reduced inflammatory cytokines such as IL-2 and IL-6, as well as a reduction in microglial activation, which contribute to an inflammatory response in neurons. Furthermore, naturally occurring cannabinoid metabolic enzymes (FAAH and MAGL) inhibit the NLRP3 inflammasome complex, which may offer significant neuroprotection. In this review, we explored the multi-targeted neuroprotective properties of phytocannabinoids and their possible modulations, which could offer significant benefits in limiting AD.
Collapse
Affiliation(s)
- Sumit S Rathod
- Department of Pharmacy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, 425405, Maharashtra, India
- Shri Vile Parle Kelavani Mandal's, Institute of Pharmacy, Dhule, Dist. Dhule, 424001, Maharashtra, India
| | - Yogeeta O Agrawal
- Department of Pharmacy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, 425405, Maharashtra, India
| |
Collapse
|
3
|
Nassif C, Kawles A, Ayala I, Minogue G, Gill NP, Shepard RA, Zouridakis A, Keszycki R, Zhang H, Mao Q, Flanagan ME, Bigio EH, Mesulam MM, Rogalski E, Geula C, Gefen T. Integrity of Neuronal Size in the Entorhinal Cortex Is a Biological Substrate of Exceptional Cognitive Aging. J Neurosci 2022; 42:8587-8594. [PMID: 36180225 PMCID: PMC9665923 DOI: 10.1523/jneurosci.0679-22.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022] Open
Abstract
Average aging is associated with a gradual decline of memory capacity. SuperAgers are humans ≥80 years of age who show exceptional episodic memory at least as good as individuals 20-30 years their junior. This study investigated whether neuronal integrity in the entorhinal cortex (ERC), an area critical for memory and selectively vulnerable to neurofibrillary degeneration, differentiated SuperAgers from cognitively healthy younger individuals, cognitively average peers ("Normal Elderly"), and individuals with amnestic mild cognitive impairment. Postmortem sections of the ERC were stained with cresyl violet to visualize neurons and immunostained with mouse monoclonal antibody PHF-1 to visualize neurofibrillary tangles. The cross-sectional area (i.e., size) of layer II and layer III/V ERC neurons were quantified. Two-thirds of total participants were female. Unbiased stereology was used to quantitate tangles in a subgroup of SuperAgers and Normal Elderly. Linear mixed-effect models were used to determine differences across groups. Quantitative measurements found that the soma size of layer II ERC neurons in postmortem brain specimens were significantly larger in SuperAgers compared with all groups (p < 0.05)-including younger individuals 20-30 years their junior (p < 0.005). SuperAgers had significantly fewer stereologically quantified Alzheimer's disease-related neurofibrillary tangles in layer II ERC than Normal Elderly (p < 0.05). This difference in tangle burden in layer II between SuperAgers and Normal Elderly suggests that tangle-bearing neurons may be prone to shrinkage during aging. The finding that SuperAgers show ERC layer II neurons that are substantially larger even compared with individuals 20-30 years younger is remarkable, suggesting that layer II ERC integrity is a biological substrate of exceptional memory in old age.SIGNIFICANCE STATEMENT Average aging is associated with a gradual decline of memory. Previous research shows that an area critical for memory, the entorhinal cortex (ERC), is susceptible to the early formation of Alzheimer's disease neuropathology, even during average (or typical) trajectories of aging. The Northwestern University SuperAging Research Program studies unique individuals known as SuperAgers, individuals ≥80 years old who show exceptional memory that is at least as good as individuals 20-30 years their junior. In this study, we show that SuperAgers harbor larger, healthier neurons in the ERC compared with their cognitively average same-aged peers, those with amnestic mild cognitive impairment, and - remarkably - even compared with individuals 20-30 years younger. We conclude that larger ERC neurons are a biological signature of the SuperAging trajectory.
Collapse
Affiliation(s)
- Caren Nassif
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Allegra Kawles
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Ivan Ayala
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Grace Minogue
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Nathan P Gill
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Robert A Shepard
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Antonia Zouridakis
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Rachel Keszycki
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Hui Zhang
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Qinwen Mao
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Eileen H Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - M-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
4
|
Incontri-Abraham D, Esparza-Salazar FJ, Ibarra A. Copolymer-1 as a potential therapy for mild cognitive impairment. Brain Cogn 2022; 162:105892. [PMID: 35841771 DOI: 10.1016/j.bandc.2022.105892] [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: 04/22/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022]
Abstract
Mild cognitive impairment (MCI) is a prodromal stage of memory impairment that may precede dementia. MCI is classified by the presence or absence of memory impairment into amnestic or non-amnestic MCI, respectively. More than 90% of patients with amnestic MCI who progress towards dementia meet criteria for Alzheimer's disease (AD). A combination of mechanisms promotes MCI, including intracellular neurofibrillary tangle formation, extracellular amyloid deposition, oxidative stress, neuronal loss, synaptodegeneration, cholinergic dysfunction, cerebrovascular disease, and neuroinflammation. However, emerging evidence indicates that neuroinflammation plays an important role in the pathogenesis of cognitive impairment. Unfortunately, there are currently no Food and Drug Administration (FDA)-approved drugs for MCI. Copolymer-1 (Cop-1), also known as glatiramer acetate, is a synthetic polypeptide of four amino acids approved by the FDA for the treatment of relapsing-remitting multiple sclerosis. Cop-1 therapeutic effect is attributed to immunomodulation, promoting a switch from proinflammatory to anti-inflammatory phenotype. In addition to its anti-inflammatory properties, it stimulates brain-derived neurotrophic factor (BDNF) secretion, a neurotrophin involved in neurogenesis and the generation of hippocampal long-term potentials. Moreover, BDNF levels are significantly decreased in patients with cognitive impairment. Therefore, Cop-1 immunization might promote synaptic plasticity and memory consolidation by increasing BDNF production in patients with MCI.
Collapse
Affiliation(s)
- Diego Incontri-Abraham
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac No. 46, Col. Lomas Anáhuac, Huixquilucan, CP 52786, Edo. de México, Mexico
| | - Felipe J Esparza-Salazar
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac No. 46, Col. Lomas Anáhuac, Huixquilucan, CP 52786, Edo. de México, Mexico
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac No. 46, Col. Lomas Anáhuac, Huixquilucan, CP 52786, Edo. de México, Mexico.
| |
Collapse
|
5
|
Martínez-Serra R, Alonso-Nanclares L, Cho K, Giese KP. Emerging insights into synapse dysregulation in Alzheimer's disease. Brain Commun 2022; 4:fcac083. [PMID: 35652120 PMCID: PMC9149787 DOI: 10.1093/braincomms/fcac083] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/15/2022] [Accepted: 04/05/2022] [Indexed: 11/14/2022] Open
Abstract
Alzheimer's disease is the leading cause of dementia and a growing worldwide problem, with its incidence expected to increase in the coming years. Since synapse loss is a major pathology and is correlated with symptoms in Alzheimer's disease, synapse dysfunction and loss may underlie pathophysiology. In this context, this review focuses on emerging insights into synaptic changes at the ultrastructural level. The three-dimensional electron microscopy technique unequivocally detects all types of synapses, including multi-synapses, which are indicators of synaptic connectivity between neurons. In recent years it has become feasible to perform sophisticated three-dimensional electron microscopy analyses on post-mortem human Alzheimer's disease brain as tissue preservation and electron microscopy techniques have improved. This ultrastructural analysis found that synapse loss does not always precede neuronal loss, as long believed. For instance, in the transentorhinal cortex and area CA1 of the hippocampus, synapse loss does not precede neuronal loss. However, in the entorhinal cortex, synapse loss precedes neuronal loss. Moreover, the ultrastructural analysis provides details about synapse morphology. For example, changes in excitatory synapses' post-synaptic densities, with fragmented postsynaptic densities increasing at the expense of perforated synapses, are seen in Alzheimer's disease brain. Further, multi-synapses also appear to be altered in Alzheimer's disease by doubling the abundance of multi-innervated spines in the transentorhinal cortex of Alzheimer's disease brain. Collectively, these recent ultrastructural analyses highlight distinct synaptic phenotypes in different Alzheimer's disease brain regions and broaden the understanding of synapse alterations, which may unravel some new therapeutic targets.
Collapse
Affiliation(s)
- Raquel Martínez-Serra
- Department of Basic and Clinical Neuroscience,
Institute of Psychiatry, Psychology and Neuroscience, King’s College
London, London SE5 9NU, UK
| | - Lidia Alonso-Nanclares
- Instituto Cajal (CSIC - Consejo Superior de
Investigaciones Científicas), Avda. Doctor Arce 37, 28002
Madrid, Spain
- Laboratorio Cajal de Circuitos Corticales (CTB),
Universidad Politécnica de Madrid, Campus de Montegancedo
s/n, Pozuelo de Alarcón 28223, Madrid, Spain
| | - Kwangwook Cho
- Department of Basic and Clinical Neuroscience,
Institute of Psychiatry, Psychology and Neuroscience, King’s College
London, London SE5 9NU, UK
- UK-Dementia Research Institute at King’s
College London, London SE5 9NU, UK
| | - K. Peter Giese
- Department of Basic and Clinical Neuroscience,
Institute of Psychiatry, Psychology and Neuroscience, King’s College
London, London SE5 9NU, UK
| |
Collapse
|
6
|
Abstract
Neurexin-3 is primarily localized in the presynaptic membrane and forms complexes with various ligands located in the postsynaptic membrane. Neurexin-3 has important roles in synapse development and synapse functions. Neurexin-3 mediates excitatory presynaptic differentiation by interacting with leucine-rich-repeat transmembrane neuronal proteins. Meanwhile, neurexin-3 modulates the expression of presynaptic α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors and γ-aminobutyric acid A receptors by interacting with neuroligins at excitatory and inhibitory synapses. Numerous studies have documented the potential contribution of neurexin-3 to neurodegenerative and neuropsychiatric disorders, such as Alzheimer's disease, addiction behaviors, and other diseases, which raises hopes that understanding the mechanisms of neurexin-3 may hold the key to developing new strategies for related illnesses. This review comprehensively covers the literature to provide current knowledge of the structure, function, and clinical role of neurexin-3.
Collapse
|
7
|
Das SR, Lyu X, Duong MT, Xie L, McCollum L, de Flores R, DiCalogero M, Irwin DJ, Dickerson BC, Nasrallah IM, Yushkevich PA, Wolk DA. Tau-Atrophy Variability Reveals Phenotypic Heterogeneity in Alzheimer's Disease. Ann Neurol 2021; 90:751-762. [PMID: 34617306 PMCID: PMC8841129 DOI: 10.1002/ana.26233] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Tau neurofibrillary tangles (T) are the primary driver of downstream neurodegeneration (N) and subsequent cognitive impairment in Alzheimer's disease (AD). However, there is substantial variability in the T-N relationship - manifested in higher or lower atrophy than expected for level of tau in a given brain region. The goal of this study was to determine if region-based quantitation of this variability allows for identification of underlying modulatory factors, including polypathology. METHODS Cortical thickness (N) and 18 F-Flortaucipir SUVR (T) were computed in 104 gray matter regions from a cohort of cognitively-impaired, amyloid-positive (A+) individuals. Region-specific residuals from a robust linear fit between SUVR and cortical thickness were computed as a surrogate for T-N mismatch. A summary T-N mismatch metric defined using residuals were correlated with demographic and imaging-based modulatory factors, and to partition the cohort into data-driven subgroups. RESULTS The summary T-N mismatch metric correlated with underlying factors such as age and burden of white matter hyperintensity lesions. Data-driven subgroups based on clustering of residuals appear to represent different biologically relevant phenotypes, with groups showing distinct spatial patterns of higher or lower atrophy than expected. INTERPRETATION These data support the notion that a measure of deviation from a normative relationship between tau burden and neurodegeneration across brain regions in individuals on the AD continuum captures variability due to multiple underlying factors, and can reveal phenotypes, which if validated, may help identify possible contributors to neurodegeneration in addition to tau, which may ultimately be useful for cohort selection in clinical trials. ANN NEUROL 2021;90:751-762.
Collapse
Affiliation(s)
- Sandhitsu R Das
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Xueying Lyu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Tran Duong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Long Xie
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren McCollum
- Department of Medicine, University of Tennessee, Knoxville, TN, USA
| | - Robin de Flores
- Université de Caen Normandie, INSERM UMRS U1237, Caen, France
| | - Michael DiCalogero
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - David J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ilya M Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A Yushkevich
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Kyrtata N, Emsley HCA, Sparasci O, Parkes LM, Dickie BR. A Systematic Review of Glucose Transport Alterations in Alzheimer's Disease. Front Neurosci 2021; 15:626636. [PMID: 34093108 PMCID: PMC8173065 DOI: 10.3389/fnins.2021.626636] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction: Alzheimer's disease (AD) is characterized by cerebral glucose hypometabolism. Hypometabolism may be partly due to reduced glucose transport at the blood-brain barrier (BBB) and across astrocytic and neuronal cell membranes. Glucose transporters (GLUTs) are integral membrane proteins responsible for moving glucose from the bloodstream to parenchymal cells where it is metabolized, and evidence indicates vascular and non-vascular GLUTs are altered in AD brains, a process which could starve the brain of glucose and accelerate cognitive decline. Here we review the literature on glucose transport alterations in AD from human and rodent studies. Methods: Literature published between 1st January 1946 and 1st November 2020 within EMBASE and MEDLINE databases was searched for the terms "glucose transporters" AND "Alzheimer's disease". Human and rodent studies were included while reviews, letters, and in-vitro studies were excluded. Results: Forty-three studies fitting the inclusion criteria were identified, covering human (23 studies) and rodent (20 studies). Post-mortem studies showed consistent reductions in GLUT1 and GLUT3 in the hippocampus and cortex of AD brains, areas of the brain closely associated with AD pathology. Tracer studies in rodent models of AD and human AD also exhibit reduced uptake of glucose and glucose-analogs into the brain, supporting these findings. Longitudinal rodent studies clearly indicate that changes in GLUT1 and GLUT3 only occur after amyloid-β pathology is present, and several studies indicate amyloid-β itself may be responsible for GLUT changes. Furthermore, evidence from human and rodent studies suggest GLUT depletion has severe effects on brain function. A small number of studies show GLUT2 and GLUT12 are increased in AD. Anti-diabetic medications improved glucose transport capacity in AD subjects. Conclusions: GLUT1 and GLUT3 are reduced in hippocampal and cortical regions in patients and rodent models of AD, and may be caused by high levels of amyloid-β in these regions. GLUT3 reductions appear to precede the onset of clinical symptoms. GLUT2 and GLUT12 appear to increase and may have a compensatory role. Repurposing anti-diabetic drugs to modify glucose transport shows promising results in human studies of AD.
Collapse
Affiliation(s)
- Natalia Kyrtata
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- University Hospitals of Morecambe Bay NHS Foundation Trust, Lancaster, United Kingdom
| | - Hedley C. A. Emsley
- Lancaster Medical School, Lancaster University, Lancaster, United Kingdom
- Department of Neurology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom
| | - Oli Sparasci
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Greater Manchester Mental Health NHS Foundation Trust, Manchester, United Kingdom
| | - Laura M. Parkes
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Ben R. Dickie
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
9
|
Haytural H, Jordà-Siquier T, Winblad B, Mulle C, Tjernberg LO, Granholm AC, Frykman S, Barthet G. Distinctive alteration of presynaptic proteins in the outer molecular layer of the dentate gyrus in Alzheimer's disease. Brain Commun 2021; 3:fcab079. [PMID: 34013204 PMCID: PMC8117432 DOI: 10.1093/braincomms/fcab079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/15/2021] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Synaptic degeneration has been reported as one of the best pathological correlates of cognitive deficits in Alzheimer's disease. However, the location of these synaptic alterations within hippocampal sub-regions, the vulnerability of the presynaptic versus postsynaptic compartments, and the biological mechanisms for these impairments remain unknown. Here, we performed immunofluorescence labelling of different synaptic proteins in fixed and paraffin-embedded human hippocampal sections and report reduced levels of several presynaptic proteins of the neurotransmitter release machinery (complexin-1, syntaxin-1A, synaptotagmin-1 and synaptogyrin-1) in Alzheimer's disease cases. The deficit was restricted to the outer molecular layer of the dentate gyrus, whereas other hippocampal sub-fields were preserved. Interestingly, standard markers of postsynaptic densities (SH3 and multiple ankyrin repeat domains protein 2) and dendrites (microtubule-associated protein 2) were unaltered, as well as the relative number of granule cells in the dentate gyrus, indicating that the deficit is preferentially presynaptic. Notably, staining for the axonal components, myelin basic protein, SMI-312 and Tau, was unaffected, suggesting that the local presynaptic impairment does not result from axonal loss or alterations of structural proteins of axons. There was no correlation between the reduction in presynaptic proteins in the outer molecular layer and the extent of the amyloid load or of the dystrophic neurites expressing phosphorylated forms of Tau. Altogether, this study highlights the distinctive vulnerability of the outer molecular layer of the dentate gyrus and supports the notion of presynaptic failure in Alzheimer's disease.
Collapse
Affiliation(s)
- Hazal Haytural
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 64 Solna, Sweden
| | - Tomàs Jordà-Siquier
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 64 Solna, Sweden
- Karolinska University Hospital, Theme Aging, 141 86 Huddinge, Sweden
| | - Christophe Mulle
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 64 Solna, Sweden
| | - Ann-Charlotte Granholm
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 64 Solna, Sweden
- Knoebel Institute for Healthy Aging, University of Denver, Denver 80208, CO, USA
| | - Susanne Frykman
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 64 Solna, Sweden
| | - Gaël Barthet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| |
Collapse
|
10
|
Domínguez-Álvaro M, Montero-Crespo M, Blazquez-Llorca L, Plaza-Alonso S, Cano-Astorga N, DeFelipe J, Alonso-Nanclares L. 3D Analysis of the Synaptic Organization in the Entorhinal Cortex in Alzheimer's Disease. eNeuro 2021; 8:ENEURO.0504-20.2021. [PMID: 34039651 PMCID: PMC8225407 DOI: 10.1523/eneuro.0504-20.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/26/2021] [Accepted: 04/16/2021] [Indexed: 01/01/2023] Open
Abstract
The entorhinal cortex (EC) is especially vulnerable in the early stages of Alzheimer's disease (AD). In particular, cognitive deficits have been linked to alterations in the upper layers of EC. In the present report, we examined Layers II and III from eight human brain autopsies (four subjects with no recorded neurologic alterations and four AD cases). We used stereological methods to assess cortical atrophy of the EC and possible changes in the volume occupied by different cortical elements (neuronal and glial cell bodies; blood vessels; and neuropil). We performed 3D ultrastructural analyses of synapses using focused ion beam/scanning electron microscopy (FIB/SEM) to examine possible alterations related to AD. At the light microscope level, we found a significantly lower volume fraction occupied by neuronal bodies in Layer III and a higher volume fraction occupied by glial cell bodies in Layer II in AD cases. At the ultrastructural level, we observed that (1) there was a significantly lower synaptic density in both layers in AD cases; (2) synapses were larger and more complex in Layer II in AD cases; and (3) there was a greater proportion of small and simple synapses in Layer III in AD cases than in control individuals. These structural differences may play a role in the anatomic basis for the impairment of cognitive functions in AD.
Collapse
Affiliation(s)
- M Domínguez-Álvaro
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
| | - M Montero-Crespo
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
| | - L Blazquez-Llorca
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
- Sección Departamental de Anatomía y Embriología (Veterinaria), Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - S Plaza-Alonso
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
| | - N Cano-Astorga
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
| | - J DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
| | - L Alonso-Nanclares
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
| |
Collapse
|
11
|
Montero-Crespo M, Domínguez-Álvaro M, Alonso-Nanclares L, DeFelipe J, Blazquez-Llorca L. Three-dimensional analysis of synaptic organization in the hippocampal CA1 field in Alzheimer's disease. Brain 2021; 144:553-573. [PMID: 33324984 PMCID: PMC8240746 DOI: 10.1093/brain/awaa406] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/07/2020] [Accepted: 09/20/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease is the most common form of dementia, characterized by a persistent and progressive impairment of cognitive functions. Alzheimer's disease is typically associated with extracellular deposits of amyloid-β peptide and accumulation of abnormally phosphorylated tau protein inside neurons (amyloid-β and neurofibrillary pathologies). It has been proposed that these pathologies cause neuronal degeneration and synaptic alterations, which are thought to constitute the major neurobiological basis of cognitive dysfunction in Alzheimer's disease. The hippocampal formation is especially vulnerable in the early stages of Alzheimer's disease. However, the vast majority of electron microscopy studies have been performed in animal models. In the present study, we performed an extensive 3D study of the neuropil to investigate the synaptic organization in the stratum pyramidale and radiatum in the CA1 field of Alzheimer's disease cases with different stages of the disease, using focused ion beam/scanning electron microscopy (FIB/SEM). In cases with early stages of Alzheimer's disease, the synapse morphology looks normal and we observed no significant differences between control and Alzheimer's disease cases regarding the synaptic density, the ratio of excitatory and inhibitory synapses, or the spatial distribution of synapses. However, differences in the distribution of postsynaptic targets and synaptic shapes were found. Furthermore, a lower proportion of larger excitatory synapses in both strata were found in Alzheimer's disease cases. Individuals in late stages of the disease suffered the most severe synaptic alterations, including a decrease in synaptic density and morphological alterations of the remaining synapses. Since Alzheimer's disease cases show cortical atrophy, our data indicate a reduction in the total number (but not the density) of synapses at early stages of the disease, with this reduction being much more accentuated in subjects with late stages of Alzheimer's disease. The observed synaptic alterations may represent a structural basis for the progressive learning and memory dysfunctions seen in Alzheimer's disease cases.
Collapse
Affiliation(s)
- Marta Montero-Crespo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Av. Doctor Arce, 37, 28002 Madrid, Spain
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Marta Domínguez-Álvaro
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Lidia Alonso-Nanclares
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Av. Doctor Arce, 37, 28002 Madrid, Spain
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, c/Valderrebollo, 5, 28031 Madrid, Spain
| | - Javier DeFelipe
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Av. Doctor Arce, 37, 28002 Madrid, Spain
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, c/Valderrebollo, 5, 28031 Madrid, Spain
| | - Lidia Blazquez-Llorca
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, c/Valderrebollo, 5, 28031 Madrid, Spain
- Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), c/Juan del Rosal, 10, 28040 Madrid, Spain
- Sección Departamental de Anatomía y Embriología (Veterinaria), Facultad de Veterinaria, Universidad Complutense de Madrid, Av. Puerta de Hierro, s/n, 28040 Madrid, Spain
| |
Collapse
|
12
|
Wu Z, Liu X, Cheng L, Ye K. Delta-secretase triggers Alzheimer's disease pathologies in wild-type hAPP/hMAPT double transgenic mice. Cell Death Dis 2020; 11:1058. [PMID: 33311478 PMCID: PMC7733592 DOI: 10.1038/s41419-020-03270-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease with multifactorial pathologies including Aβ containing senile plaques and neurofibrillary tangles (NFT) consisted of aggregated Tau. Most of the AD patients are sporadic and the familial mutation hereditary patients are composed only 1% of all cases. However, the current AD mouse models employ mutated APP, PS1, or even Tau mutant, in order to display a portion of AD pathologies. Delta-secretase (legumain, or asparaginyl endopeptidase, AEP) simultaneously cleaves both APP and Tau and augments Aβ production and Tau hyperphosphorylation and aggregation, contributing to AD pathogenesis. Here we show that δ-secretase is sufficient to promote prominent AD pathologies in wild-type hAPP/hMAPT double transgenic mice. We crossed hAPP l5 mice and hMAPT mice to generate double transgenic mouse model carrying both human wild-type APP and Tau. Compared to the single transgenic parents, these double transgenic mice demonstrated AD-related pathologies in one-year-old hAPP/hMAPT mice. Notably, overexpression of δ-secretase in hAPP/hMAPT double-transgenic mice evidently accelerated enormous senile plaques and NFT, associated with prominent synaptic defects and cognitive deficits. Hence, δ-secretase facilitates AD pathogenesis independent of any patient-derived mutation.
Collapse
Affiliation(s)
- Zhourui Wu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Spine, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, 200065, China
- Key Laboratory of spine and spinal cord injury repair and regeneration, Ministry of Education of the People's Republic of China, Shanghai, 200072, China
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, 200065, China.
- Key Laboratory of spine and spinal cord injury repair and regeneration, Ministry of Education of the People's Republic of China, Shanghai, 200072, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
13
|
Honer WG, Ramos-Miguel A, Alamri J, Sawada K, Barr AM, Schneider JA, Bennett DA. The synaptic pathology of cognitive life
. DIALOGUES IN CLINICAL NEUROSCIENCE 2020; 21:271-279. [PMID: 31749651 PMCID: PMC6829169 DOI: 10.31887/dcns.2019.21.3/whoner] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Prospective, community-based studies allow evaluation of associations between
cognitive functioning and synaptic measures, controlled for age-related pathologies.
Findings from >400 community-based participants are reviewed. Levels of two
presynaptic proteins, complexin-I (inhibitory terminals), and complexin-II (excitatory
terminals) contributed to cognitive variation from normal to dementia. Adding the amount
of protein-protein interaction between two others, synaptosome-associated protein-25 and
syntaxin, explained 6% of overall variance. The presynaptic protein Munc18-1 long
variant was localized to inhibitory terminals, and like complexin-I, was positively
associated with cognition. Associations depended on Braak stage, with the level of
complexin-I contributing nearly 15% to cognitive variation in stages 0-II, while
complexin-II contributed 7% in stages V-VI. Non-denaturing gels identified multiple
soluble N-ethylmaleimide-sensitive factor attachment protein receptor protein-protein
(SNARE) complexes in frontal and in temporal lobes, making specific contributions to
cognitive functions. Multiple mechanisms of presynaptic plasticity contribute to
cognitive function during aging.
Collapse
Affiliation(s)
- William G Honer
- Departments of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alfredo Ramos-Miguel
- Department of Pharmacology, University of the Basque Country, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Jehan Alamri
- Departments of Anaesthesia, Pharmacology & Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Alasdair M Barr
- Departments of Anaesthesia, Pharmacology & Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, US
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, US
| |
Collapse
|
14
|
Corsetti V, Borreca A, Latina V, Giacovazzo G, Pignataro A, Krashia P, Natale F, Cocco S, Rinaudo M, Malerba F, Florio R, Ciarapica R, Coccurello R, D’Amelio M, Ammassari-Teule M, Grassi C, Calissano P, Amadoro G. Passive immunotherapy for N-truncated tau ameliorates the cognitive deficits in two mouse Alzheimer's disease models. Brain Commun 2020; 2:fcaa039. [PMID: 32954296 PMCID: PMC7425324 DOI: 10.1093/braincomms/fcaa039] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/05/2020] [Accepted: 02/12/2020] [Indexed: 12/12/2022] Open
Abstract
Clinical and neuropathological studies have shown that tau pathology better correlates with the severity of dementia than amyloid plaque burden, making tau an attractive target for the cure of Alzheimer's disease. We have explored whether passive immunization with the 12A12 monoclonal antibody (26-36aa of tau protein) could improve the Alzheimer's disease phenotype of two well-established mouse models, Tg2576 and 3xTg mice. 12A12 is a cleavage-specific monoclonal antibody which selectively binds the pathologically relevant neurotoxic NH226-230 fragment (i.e. NH2htau) of tau protein without cross-reacting with its full-length physiological form(s). We found out that intravenous administration of 12A12 monoclonal antibody into symptomatic (6 months old) animals: (i) reaches the hippocampus in its biologically active (antigen-binding competent) form and successfully neutralizes its target; (ii) reduces both pathological tau and amyloid precursor protein/amyloidβ metabolisms involved in early disease-associated synaptic deterioration; (iii) improves episodic-like type of learning/memory skills in hippocampal-based novel object recognition and object place recognition behavioural tasks; (iv) restores the specific up-regulation of the activity-regulated cytoskeleton-associated protein involved in consolidation of experience-dependent synaptic plasticity; (v) relieves the loss of dendritic spine connectivity in pyramidal hippocampal CA1 neurons; (vi) rescues the Alzheimer's disease-related electrophysiological deficits in hippocampal long-term potentiation at the CA3-CA1 synapses; and (vii) mitigates the neuroinflammatory response (reactive gliosis). These findings indicate that the 20-22 kDa NH2-terminal tau fragment is crucial target for Alzheimer's disease therapy and prospect immunotherapy with 12A12 monoclonal antibody as safe (normal tau-preserving), beneficial approach in contrasting the early Amyloidβ-dependent and independent neuropathological and cognitive alterations in affected subjects.
Collapse
Affiliation(s)
| | - Antonella Borreca
- Humanitas University Laboratory of Pharmacology and Brain Pathology, Neuro Center, 20089 Milan, Italy
- Institute of Neuroscience, 20129 Milan, Italy
| | | | | | | | - Paraskevi Krashia
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Department of Medicine, University Campus Bio-Medico, 00128 Rome, Italy
- Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128 Rome, Italy
| | - Francesca Natale
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Sara Cocco
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Marco Rinaudo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | | | - Rita Florio
- European Brain Research Institute (EBRI), 00161 Rome, Italy
| | | | - Roberto Coccurello
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Institute for Complex Systems (ISC), CNR, 00185 Rome, Italy
| | - Marcello D’Amelio
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Department of Medicine, University Campus Bio-Medico, 00128 Rome, Italy
- Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128 Rome, Italy
| | | | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), 00161 Rome, Italy
- Institute of Translational Pharmacology (IFT)–National Research Council (CNR), 00133 Rome, Italy
| |
Collapse
|
15
|
Alterations in Synaptic Plasticity and Oxidative Stress Following Long-Term Paracetamol Treatment in Rat Brain. Neurotox Res 2019; 37:455-468. [PMID: 31364032 DOI: 10.1007/s12640-019-00090-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 07/03/2019] [Accepted: 07/12/2019] [Indexed: 10/26/2022]
Abstract
Several studies have recently revealed that cognitive function can be affected by paracetamol (APAP) treatment. However, the exact impact of this drug treatment on learning and memory has not been clarified. This study aimed to investigate the effect of APAP treatment on the alteration of synapses and oxidative stress in the rat frontal cortex and hippocampus. APAP at a dose of 200 mg/kg bw was fed to adult male Wistar rats through either acute (n = 10), 15-day (n = 10), or 30-day (n = 10) treatment regimens. The synaptic ultrastructure and proteins, synaptophysin (SYP) and postsynaptic density-95 (PSD-95), were monitored. The amount of protein carbonyl oxidation (PCO) and glutathione (GSH) levels were examined. Our results demonstrated that acute treatment with APAP had no effect on synapses and oxidative stress. However, the synapses obtained from rats with 15-day APAP treatment showed a marked shortening of active zones and widening of the synaptic cleft. Decrement of SYP and PSD-95 proteins were demonstrated in these rats as well. With 30-day APAP treatment, the alteration of the synaptic ultrastructure and proteins was more evident. Moreover, the depletion of GSH and the elevation of PCO levels were demonstrated in the rats treated with APAP for 30 days. These results suggest that long-term APAP treatment can induce synaptic degeneration in the hippocampus and frontal cortex. The increase in oxidative stress in these brain areas may be due to the deleterious effect of this drug.
Collapse
|
16
|
Chen XQ, Mobley WC. Alzheimer Disease Pathogenesis: Insights From Molecular and Cellular Biology Studies of Oligomeric Aβ and Tau Species. Front Neurosci 2019; 13:659. [PMID: 31293377 PMCID: PMC6598402 DOI: 10.3389/fnins.2019.00659] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/07/2019] [Indexed: 01/08/2023] Open
Abstract
Alzheimer disease (AD) represents an oncoming epidemic that without an effective treatment promises to exact extraordinary human and financial burdens. Studies of pathogenesis are essential for defining targets for discovering disease-modifying treatments. Past studies of AD neuropathology provided valuable, albeit limited, insights. Nevertheless, building on these findings, recent studies have provided an increasingly rich harvest of genetic, molecular and cellular data that are creating unprecedented opportunities to both understand and treat AD. Among the most significant are those documenting the presence within the AD brain of toxic oligomeric species of Aβ and tau. Existing data support the view that such species can propagate and spread within neural circuits. To place these findings in context we first review the genetics and neuropathology of AD, including AD in Down syndrome (AD-DS). We detail studies that support the existence of toxic oligomeric species while noting the significant unanswered questions concerning their precise structures, the means by which they spread and undergo amplification and how they induce neuronal dysfunction and degeneration. We conclude by offering a speculative synthesis for how oligomers of Aβ and tau initiate and drive pathogenesis. While 100 years after Alzheimer's first report there is much still to learn about pathogenesis and the discovery of disease-modifying treatments, the application of new concepts and sophisticated new tools are poised to deliver important advances for combatting AD.
Collapse
Affiliation(s)
- Xu-Qiao Chen
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - William C. Mobley
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
17
|
Sui Y, Zhang Y, Dong C, Xu B, Sun X. The small molecular CCR3 antagonist YM344031 attenuates neurodegenerative pathologies and improves learning and memory performance in a mouse model of Alzheimer's disease. Brain Res 2019; 1719:1-10. [PMID: 31121157 DOI: 10.1016/j.brainres.2019.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/03/2019] [Accepted: 05/19/2019] [Indexed: 12/13/2022]
Abstract
The chemokine C-C receptor 3 (CCR3) plays a role in the pathogenesis of Alzheimer's disease (AD). Based on our previous observations that deletion of CCR3 prevented neurodegenerative pathologies in amyloid precursor protein/presenilin 1 (APP/PS1) double-transgenic mice, we hypothesize that CCR3 antagonists may provide therapeutic benefits to AD. To this end, we examined the effect of the brain-penetrable CCR3 antagonist, YM344031, on AD-related pathologies in APP/PS1 double transgenic mice. Treatment of 10-month-old APP/PS1 double-transgenic mice with YM344031 (50 mg/kg, b.i.d.) for two months resulted in dramatic decreases in β-amyloid deposition, tau hyperphosphorylation and synaptic loss in the forebrain, significant attenuation of microgliosis and astrogliosis, and marked improvement of spatial learning and memory performance compared with the vehicle-treated mice. These results support CCR3 antagonism as a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Yi Sui
- Department of Neurology, Shenyang First People's Hospital, Shenyang Brain Institute, Shenyang Medical College Affiliated Shenyang Brain Hospital, Shenyang 110047, China
| | - Yao Zhang
- Department of Neurology, Shenyang First People's Hospital, Shenyang Brain Institute, Shenyang Medical College Affiliated Shenyang Brain Hospital, Shenyang 110047, China
| | - Chunyao Dong
- Department of Neurology, Shenyang First People's Hospital, Shenyang Brain Institute, Shenyang Medical College Affiliated Shenyang Brain Hospital, Shenyang 110047, China
| | - Bing Xu
- Department of Neurology, Shenyang First People's Hospital, Shenyang Brain Institute, Shenyang Medical College Affiliated Shenyang Brain Hospital, Shenyang 110047, China
| | - Xiaohong Sun
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang 110004, China.
| |
Collapse
|
18
|
Ramos-Miguel A, Jones AA, Sawada K, Barr AM, Bayer TA, Falkai P, Leurgans SE, Schneider JA, Bennett DA, Honer WG. Frontotemporal dysregulation of the SNARE protein interactome is associated with faster cognitive decline in old age. Neurobiol Dis 2018; 114:31-44. [PMID: 29496544 PMCID: PMC6483375 DOI: 10.1016/j.nbd.2018.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 02/07/2018] [Accepted: 02/21/2018] [Indexed: 12/29/2022] Open
Abstract
The molecular underpinnings associated with cognitive reserve remain poorly understood. Because animal models fail to fully recapitulate the complexity of human brain aging, postmortem studies from well-designed cohorts are crucial to unmask mechanisms conferring cognitive resistance against cumulative neuropathologies. We tested the hypothesis that functionality of the SNARE protein interactome might be an important resilience factor preserving cognitive abilities in old age. Cognition was assessed annually in participants from the Rush "Memory and Aging Project" (MAP), a community-dwelling cohort representative of the overall aging population. Associations between cognition and postmortem neurochemical data were evaluated in functional assays quantifying various species of the SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) machinery in samples from the inferior temporal (IT, n = 154) and middle-frontal (MF, n = 174) gyri. Using blue-native gel electrophoresis, we isolated and quantified several types of complexes containing the three SNARE proteins (syntaxin-1, SNAP25, VAMP), as well as the GABAergic/glutamatergic selectively expressed complexins-I/II (CPLX1/2), in brain tissue homogenates and reconstitution assays with recombinant proteins. Multivariate analyses revealed significant associations between IT and MF neurochemical data (SNARE proteins and/or complexes), and multiple age-related neuropathologies, as well as with multiple cognitive domains of MAP participants. Controlling for demographic variables, neuropathologic indices and total synapse density, we found that temporal 150-kDa SNARE species (representative of pan-synaptic functionality) and frontal CPLX1/CPLX2 ratio of 500-kDa heteromeric species (representative of inhibitory/excitatory input functionality) were, among all the immunocharacterized complexes, the strongest predictors of cognitive function nearest death. Interestingly, these two neurochemical variables were associated with different cognitive domains. In addition, linear mixed effect models of global cognitive decline estimated that both 150-kDa SNARE levels and CPLX1/CPLX2 ratio were associated with better cognition and less decline over time. The results are consistent with previous studies reporting that synapse dysfunction (i.e. dysplasticity) may be initiated early, and relatively independent of neuropathology-driven synapse loss. Frontotemporal dysregulation of the GABAergic/glutamatergic stimuli might be a target for future drug development.
Collapse
Affiliation(s)
- Alfredo Ramos-Miguel
- BC Mental Health and Addictions Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 2A1, Canada
| | - Andrea A Jones
- BC Mental Health and Addictions Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 2A1, Canada
| | - Ken Sawada
- Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi 783-8505, Japan
| | - Alasdair M Barr
- BC Mental Health and Addictions Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Thomas A Bayer
- Department of Psychiatry, University Medicine Goettingen, von-Siebold-Strasse 5, D-37075 Goettingen, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Nussbaumstrasse 7, D-80336 Munich, Germany
| | - Sue E Leurgans
- Rush Alzheimer's disease Center, Rush University Medical Center, 600 S. Paulina Street, Chicago, IL 60612, United States
| | - Julie A Schneider
- Rush Alzheimer's disease Center, Rush University Medical Center, 600 S. Paulina Street, Chicago, IL 60612, United States
| | - David A Bennett
- Rush Alzheimer's disease Center, Rush University Medical Center, 600 S. Paulina Street, Chicago, IL 60612, United States
| | - William G Honer
- BC Mental Health and Addictions Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 2A1, Canada.
| |
Collapse
|
19
|
AD-Related N-Terminal Truncated Tau Is Sufficient to Recapitulate In Vivo the Early Perturbations of Human Neuropathology: Implications for Immunotherapy. Mol Neurobiol 2018; 55:8124-8153. [PMID: 29508283 DOI: 10.1007/s12035-018-0974-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/19/2018] [Indexed: 01/08/2023]
Abstract
The NH2tau 26-44 aa (i.e., NH2htau) is the minimal biologically active moiety of longer 20-22-kDa NH2-truncated form of human tau-a neurotoxic fragment mapping between 26 and 230 amino acids of full-length protein (htau40)-which is detectable in presynaptic terminals and peripheral CSF from patients suffering from AD and other non-AD neurodegenerative diseases. Nevertheless, whether its exogenous administration in healthy nontransgenic mice is able to elicit a neuropathological phenotype resembling human tauopathies has not been yet investigated. We explored the in vivo effects evoked by subchronic intracerebroventricular (i.c.v.) infusion of NH2htau or its reverse counterpart into two lines of young (2-month-old) wild-type mice (C57BL/6 and B6SJL). Six days after its accumulation into hippocampal parenchyma, significant impairment in memory/learning performance was detected in NH2htau-treated group in association with reduced synaptic connectivity and neuroinflammatory response. Compromised short-term plasticity in paired-pulse facilitation paradigm (PPF) was detected in the CA3/CA1 synapses from NH2htau-impaired animals along with downregulation in calcineurin (CaN)-stimulated pCREB/c-Fos pathway(s). Importantly, these behavioral, synaptotoxic, and neuropathological effects were independent from the genetic background, occurred prior to frank neuronal loss, and were specific because no alterations were detected in the control group infused with its reverse counterpart. Finally, a 2.0-kDa peptide which biochemically and immunologically resembles the injected NH2htau was endogenously detected in vivo, being present in hippocampal synaptosomal preparations from AD subjects. Given that the identification of the neurotoxic tau species is mandatory to develop a more effective tau-based immunological approach, our evidence can have important translational implications for cure of human tauopathies.
Collapse
|
20
|
Domínguez-Álvaro M, Montero-Crespo M, Blazquez-Llorca L, Insausti R, DeFelipe J, Alonso-Nanclares L. Three-dimensional analysis of synapses in the transentorhinal cortex of Alzheimer's disease patients. Acta Neuropathol Commun 2018; 6:20. [PMID: 29499755 PMCID: PMC5834884 DOI: 10.1186/s40478-018-0520-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 02/17/2018] [Indexed: 12/11/2022] Open
Abstract
Synaptic dysfunction or loss in early stages of Alzheimer’s disease (AD) is thought to be a major structural correlate of cognitive dysfunction. Early loss of episodic memory, which occurs at the early stage of AD, is closely associated with the progressive degeneration of medial temporal lobe (MTL) structures of which the transentorhinal cortex (TEC) is the first affected area. However, no ultrastructural studies have been performed in this region in human brain samples from AD patients. In the present study, we have performed a detailed three-dimensional (3D) ultrastructural analysis using focused ion beam/scanning electron microscopy (FIB/SEM) to investigate possible synaptic alterations in the TEC of patients with AD. Surprisingly, the analysis of the density, morphological features and spatial distribution of synapses in the neuropil showed no significant differences between AD and control samples. However, light microscopy studies showed that cortical thickness of the TEC was severely reduced in AD samples, but there were no changes in the volume occupied by neuronal and glial cell bodies, blood vessels, and neuropil. Thus, the present results indicate that there is a dramatic loss of absolute number of synapses, while the morphology of synaptic junctions and synaptic spatial distribution are maintained. How these changes affect cognitive impairment in AD remains to be elucidated.
Collapse
|
21
|
Pan W, Han S, Kang L, Li S, Du J, Cui H. Effects of dihydrotestosterone on synaptic plasticity of the hippocampus in mild cognitive impairment male SAMP8 mice. Exp Ther Med 2016; 12:1455-1463. [PMID: 27588067 PMCID: PMC4997989 DOI: 10.3892/etm.2016.3470] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 06/21/2016] [Indexed: 01/03/2023] Open
Abstract
The current study focused on how dihydrotestosterone (DHT) regulates synaptic plasticity in the hippocampus of mild cognitive impairment male senescence-accelerated mouse prone 8 (SAMP8) mice. Five-month-old SAMP8 mice were divided into the control, castrated and castrated-DHT groups, in which the mice were castrated and treated with physiological doses of DHT for a period of 2 months. To determine the regulatory mechanisms of DHT in the cognitive capacity, the effects of DHT on the morphology of the synapse and the expression of synaptic marker proteins in the hippocampus were investigated using immunohistochemistry, qPCR and western blot analysis. The results showed that the expression of cAMP-response element binding protein (CREB), postsynaptic density protein 95 (PSD95), synaptophysin (SYN) and developmentally regulated brain protein (Drebrin) was reduced in the castrated group compared to the control group. However, DHT promoted the expression of CREB, PSD95, SYN and Drebrin in the hippocampus of the castrated-DHT group. Thus, androgen depletion impaired the synaptic plasticity in the hippocampus of SAMP8 and accelerated the development of Alzheimer's disease (AD)-like neuropathology, suggesting that a similar mechanism may underlie the increased risk for AD in men with low testosterone. In addition, DHT regulated synaptic plasticity in the hippocampus of mild cognitive impairment (MCI) SAMP8 mice and delayed the progression of disease to Alzheimer's dementia. In conclusion, androgen-based hormone therapy is a potentially useful strategy for preventing the progression of MCI in aging men. Androgens enhance synaptic markers (SYN, PSD95, and Drebrin), activate CREB, modulate the fundamental biology of synaptic structure, and lead to the structural changes of plasticity in the hippocampus, all of which result in improved cognitive function.
Collapse
Affiliation(s)
- Wensen Pan
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China; Department of Respiration Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Shuo Han
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Lin Kang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Sha Li
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Juan Du
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Huixian Cui
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China; Hebei Key Laboratory for Brain Aging and Cognitive Neuroscience, Shijiazhuang, Hebei 050031, P.R. China
| |
Collapse
|
22
|
Regulation of the Postsynaptic Compartment of Excitatory Synapses by the Actin Cytoskeleton in Health and Its Disruption in Disease. Neural Plast 2016; 2016:2371970. [PMID: 27127658 PMCID: PMC4835652 DOI: 10.1155/2016/2371970] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/09/2016] [Indexed: 02/07/2023] Open
Abstract
Disruption of synaptic function at excitatory synapses is one of the earliest pathological changes seen in wide range of neurological diseases. The proper control of the segregation of neurotransmitter receptors at these synapses is directly correlated with the intact regulation of the postsynaptic cytoskeleton. In this review, we are discussing key factors that regulate the structure and dynamics of the actin cytoskeleton, the major cytoskeletal building block that supports the postsynaptic compartment. Special attention is given to the complex interplay of actin-associated proteins that are found in the synaptic specialization. We then discuss our current understanding of how disruption of these cytoskeletal elements may contribute to the pathological events observed in the nervous system under disease conditions with a particular focus on Alzheimer's disease pathology.
Collapse
|
23
|
Vandal M, White PJ, Tournissac M, Tremblay C, St-Amour I, Drouin-Ouellet J, Bousquet M, Traversy MT, Planel E, Marette A, Calon F. Impaired thermoregulation and beneficial effects of thermoneutrality in the 3×Tg-AD model of Alzheimer's disease. Neurobiol Aging 2016; 43:47-57. [PMID: 27255814 DOI: 10.1016/j.neurobiolaging.2016.03.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
The sharp rise in the incidence of Alzheimer's disease (AD) at an old age coincides with a reduction in energy metabolism and core body temperature. We found that the triple-transgenic mouse model of AD (3×Tg-AD) spontaneously develops a lower basal body temperature and is more vulnerable to a cold environment compared with age-matched controls. This was despite higher nonshivering thermogenic activity, as evidenced by brown adipose tissue norepinephrine content and uncoupling protein 1 expression. A 24-hour exposure to cold (4 °C) aggravated key neuropathologic markers of AD such as: tau phosphorylation, soluble amyloid beta concentrations, and synaptic protein loss in the cortex of 3×Tg-AD mice. Strikingly, raising the body temperature of aged 3×Tg-AD mice via exposure to a thermoneutral environment improved memory function and reduced amyloid and synaptic pathologies within a week. Our results suggest the presence of a vicious cycle between impaired thermoregulation and AD-like neuropathology, and it is proposed that correcting thermoregulatory deficits might be therapeutic in AD.
Collapse
Affiliation(s)
- Milene Vandal
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Québec, Canada
| | - Philip J White
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, NC, USA; Faculté de medicine, Université Laval, Québec, Québec, Canada; Institut universitaire de pneumologie et de cardiologie de Québec, Québec, Québec, Canada
| | - Marine Tournissac
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Québec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada
| | - Isabelle St-Amour
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Département de Recherche et Développement, Héma-Québec, Québec, Québec, Canada
| | - Janelle Drouin-Ouellet
- Faculté de medicine, Université Laval, Québec, Québec, Canada; John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Melanie Bousquet
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Québec, Canada
| | - Marie-Thérèse Traversy
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada
| | - Emmanuel Planel
- Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Faculté de medicine, Université Laval, Québec, Québec, Canada
| | - Andre Marette
- Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Québec, Canada; Faculté de medicine, Université Laval, Québec, Québec, Canada; Institut universitaire de pneumologie et de cardiologie de Québec, Québec, Québec, Canada
| | - Frederic Calon
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Québec, Canada; Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
24
|
Ramos-Miguel A, Hercher C, Beasley CL, Barr AM, Bayer TA, Falkai P, Leurgans SE, Schneider JA, Bennett DA, Honer WG. Loss of Munc18-1 long splice variant in GABAergic terminals is associated with cognitive decline and increased risk of dementia in a community sample. Mol Neurodegener 2015; 10:65. [PMID: 26628003 PMCID: PMC4667524 DOI: 10.1186/s13024-015-0061-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/24/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Presynaptic terminals contribute to cognitive reserve, balancing the effects of age-related pathologies on cognitive function in the elderly. The presynaptic protein Munc18-1, alternatively spliced into long (M18L) or short (M18S) isoforms, is a critical modulator of neurotransmission. While subtle alterations in Munc18-1 have been shown to cause severe neuropsychiatric disorders with cognitive impairment, little information is known regarding the specific roles of Munc18-1 splice variants. We first investigated functional and anatomical features evidencing the divergent roles of M18L and M18S, and then evaluated their contribution to the full range of age-related cognitive impairment in the dorsolateral prefrontal cortex of a large sample of participants from a community-based aging study, including subjects with no-(NCI, n = 90), or mild-(MCI, n = 86) cognitive impairment, or with clinical dementia (n = 132). Finally, we used APP23 mutant mice to study the association between M18L/S and the time-dependent accumulation of common Alzheimer's disease pathology. RESULTS Using isoform-specific antibodies, M18L was localized to the synaptosomal fraction, with a distribution matching lipid raft microdomains. M18S was found widely across cytosolic and synaptosomal compartments. Immunocytochemical studies identified M18L in perisomatic, GABAergic terminals, while M18S was broadly distributed in GABAergic and glutamatergic terminals. Using regression models taking into account multiple age-related pathologies, age, education and sex, global cognitive function was associated with the level of M18L (p = 0.006) but not M18S (p = 0.88). Mean M18L in dementia cases was 51 % lower than in NCI cases (p < 0.001), and each unit of M18L was associated with a lower likelihood of dementia (odds ratio = 0.68, 95 % confidence interval = 0.50-0.90, p = 0.008). In contrast, M18S balanced across clinical and pathologically diagnosed groups. M18L loss may not be caused by age-related amyloid pathology, since APP23 mice (12- and 22-months of age) had unchanged cortical levels of M18L/S compared with wild-type animals. CONCLUSIONS M18L was localized to presynaptic inhibitory terminals, and was associated with cognitive function and protection from dementia in an elderly, community-based cohort. Lower M18L in inhibitory presynaptic terminals may be an early, independent contributor to cognitive decline.
Collapse
Affiliation(s)
- Alfredo Ramos-Miguel
- Child and Family Research Institute, 938 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada. .,Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 2A1, Canada.
| | - Christa Hercher
- Child and Family Research Institute, 938 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada. .,Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 2A1, Canada.
| | - Clare L Beasley
- Child and Family Research Institute, 938 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada. .,Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 2A1, Canada.
| | - Alasdair M Barr
- Child and Family Research Institute, 938 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada. .,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Thomas A Bayer
- Department of Psychiatry, University Medicine Goettingen, von-Siebold-Strasse 5, D-37075, Goettingen, Germany.
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Nussbaumstrasse 7, D-80336, Munich, Germany.
| | - Sue E Leurgans
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600S. Paulina Street, IL, 60612, Chicago, USA.
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600S. Paulina Street, IL, 60612, Chicago, USA.
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600S. Paulina Street, IL, 60612, Chicago, USA.
| | - William G Honer
- Child and Family Research Institute, 938 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada. .,Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 2A1, Canada.
| |
Collapse
|
25
|
Jadhav S, Cubinkova V, Zimova I, Brezovakova V, Madari A, Cigankova V, Zilka N. Tau-mediated synaptic damage in Alzheimer's disease. Transl Neurosci 2015; 6:214-226. [PMID: 28123806 PMCID: PMC4936631 DOI: 10.1515/tnsci-2015-0023] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/04/2015] [Indexed: 12/16/2022] Open
Abstract
Synapses are the principal sites for chemical communication between neurons and are essential for performing the dynamic functions of the brain. In Alzheimer’s disease and related tauopathies, synapses are exposed to disease modified protein tau, which may cause the loss of synaptic contacts that culminate in dementia. In recent decades, structural, transcriptomic and proteomic studies suggest that Alzheimer’s disease represents a synaptic disorder. Tau neurofibrillary pathology and synaptic loss correlate well with cognitive impairment in these disorders. Moreover, regional distribution and the load of neurofibrillary lesions parallel the distribution of the synaptic loss. Several transgenic models of tauopathy expressing various forms of tau protein exhibit structural synaptic deficits. The pathological tau proteins cause the dysregulation of synaptic proteome and lead to the functional abnormalities of synaptic transmission. A large body of evidence suggests that tau protein plays a key role in the synaptic impairment of human tauopathies.
Collapse
Affiliation(s)
- Santosh Jadhav
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska 9, 845 10 Bratislava, Slovak Republic
| | - Veronika Cubinkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska 9, 845 10 Bratislava, Slovak Republic; Axon Neuroscience SE, Grosslingova 45, Bratislava, Slovak Republic
| | - Ivana Zimova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska 9, 845 10 Bratislava, Slovak Republic; Axon Neuroscience SE, Grosslingova 45, Bratislava, Slovak Republic
| | - Veronika Brezovakova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska 9, 845 10 Bratislava, Slovak Republic
| | - Aladar Madari
- Small animal clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, Kosice, Slovak Republic
| | - Viera Cigankova
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovak Republic
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska 9, 845 10 Bratislava, Slovak Republic; Axon Neuroscience SE, Grosslingova 45, Bratislava, Slovak Republic
| |
Collapse
|
26
|
Chang RYK, Etheridge N, Nouwens AS, Dodd PR. SWATH analysis of the synaptic proteome in Alzheimer's disease. Neurochem Int 2015; 87:1-12. [PMID: 25958317 DOI: 10.1016/j.neuint.2015.04.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 04/20/2015] [Accepted: 04/21/2015] [Indexed: 10/23/2022]
Abstract
Brain tissue from Alzheimer's disease patients exhibits synaptic degeneration in selected regions. Synaptic dysfunction occurs early in the disease and is a primary pathological target for treatment. The molecular mechanisms underlying this degeneration remain unknown. Quantifying the synaptic proteome in autopsy brain and comparing tissue from Alzheimer's disease cases and subjects with normal aging are critical to understanding the molecular mechanisms associated with Alzheimer pathology. We isolated synaptosomes from hippocampus and motor cortex so as to reduce sample complexity relative to whole-tissue homogenates. Synaptosomal extracts were subjected to strong cation exchange (SCX) fractionation to further partition sample complexity; each fraction received SWATH-based information-dependent acquisition to generate a comprehensive peptide-ion library. The expression of synaptic proteins from AD hippocampus and motor cortex was then compared between groups. A total of 2077 unique proteins were identified at a critical local false discovery rate <5%. Thirty of these, including 17 novel proteins, exhibited significant expression differences between cases and controls; these proteins are involved in cellular functions including structural maintenance, signal transduction, autophagy, oxidative stress, and proteasome activity, or they have synaptic-vesicle related or energy-related functions. Differentially expressed proteins were subjected to pathway analysis to identify protein-protein interactions. This revealed that the most perturbed molecular and cellular functions were cellular assembly and organization. Core analysis revealed RhoA signaling to be the top canonical pathway. Network analysis showed that differentially expressed proteins were related to cellular assembly and organization, and cellular function and maintenance. This is the first study to combine SCX fractionation with SWATH analysis. SWATH is a promising new technique that can greatly enhance protein identification in any proteome, and has many other benefits; however, there are limitations yet to be resolved.
Collapse
Affiliation(s)
| | - Naomi Etheridge
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia
| | - Amanda S Nouwens
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia
| | - Peter R Dodd
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia.
| |
Collapse
|
27
|
Jadhav S, Katina S, Kovac A, Kazmerova Z, Novak M, Zilka N. Truncated tau deregulates synaptic markers in rat model for human tauopathy. Front Cell Neurosci 2015; 9:24. [PMID: 25755633 PMCID: PMC4337338 DOI: 10.3389/fncel.2015.00024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/14/2015] [Indexed: 01/04/2023] Open
Abstract
Synaptic failure and neurofibrillary degeneration are two major neuropathological substrates of cognitive dysfunction in Alzheimer’s disease (AD). Only a few studies have demonstrated a direct relationship between these two AD hallmarks. To investigate tau mediated synaptic injury we used rat model of tauopathy that develops extensive neurofibrillary pathology in the cortex. Using fractionation of cortical synapses, we identified an increase in endogenous rat tau isoforms in presynaptic compartment, and their mis-sorting to the postsynaptic density (PSD). Truncated transgenic tau was distributed in both compartments exhibiting specific phospho-pattern that was characteristic for each synaptic compartment. In the presynaptic compartment, truncated tau was associated with impairment of dynamic stability of microtubules which could be responsible for reduction of synaptic vesicles. In the PSD, truncated tau lowered the levels of neurofilaments. Truncated tau also significantly decreased the synaptic levels of Aβ40 but not Aβ42. These data show that truncated tau differentially deregulates synaptic proteome in pre- and postsynaptic compartments. Importantly, we show that alteration of Aβ can arise downstream of truncated tau pathology.
Collapse
Affiliation(s)
- Santosh Jadhav
- Institute of Neuroimmunology, Slovak Academy of Sciences Bratislava, Slovak Republic
| | - Stanislav Katina
- Axon Neuroscience GmbH Bratislava, Slovak Republic ; Institute of Mathematics and Statistics, Masaryk University Brno, Czech Republic
| | - Andrej Kovac
- Axon Neuroscience GmbH Bratislava, Slovak Republic
| | - Zuzana Kazmerova
- Institute of Neuroimmunology, Slovak Academy of Sciences Bratislava, Slovak Republic
| | - Michal Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences Bratislava, Slovak Republic ; Axon Neuroscience GmbH Bratislava, Slovak Republic
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences Bratislava, Slovak Republic ; Axon Neuroscience GmbH Bratislava, Slovak Republic
| |
Collapse
|
28
|
Sindi IA, Tannenberg RK, Dodd PR. Role for the neurexin-neuroligin complex in Alzheimer's disease. Neurobiol Aging 2013; 35:746-56. [PMID: 24211009 DOI: 10.1016/j.neurobiolaging.2013.09.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 09/20/2013] [Accepted: 09/22/2013] [Indexed: 11/29/2022]
Abstract
Synaptic damage is a critical hallmark of Alzheimer's disease, and the best correlate with cognitive impairment ante mortem. Synapses, the loci of communication between neurons, are characterized by signature protein combinations arrayed at tightly apposed pre- and post-synaptic sites. The most widely studied trans-synaptic junctional complexes, which direct synaptogenesis and foster the maintenance and stability of the mature terminal, are conjunctions of presynaptic neurexins and postsynaptic neuroligins. Fluctuations in the levels of neuroligins and neurexins can sway the balance between excitatory and inhibitory neurotransmission in the brain, and could lead to damage of synapses and dendrites. This review summarizes current understanding of the roles of neurexins and neuroligins proteolytic processing in synaptic plasticity in the human brain, and outlines their possible roles in β-amyloid metabolism and function, which are central pathogenic events in Alzheimer's disease progression.
Collapse
Affiliation(s)
- Ikhlas A Sindi
- Centre for Psychiatry and Clinical Neuroscience, School of Medicine, The University of Queensland, Brisbane, Australia
| | - Rudolph K Tannenberg
- Centre for Psychiatry and Clinical Neuroscience, School of Medicine, The University of Queensland, Brisbane, Australia; School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Peter R Dodd
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
29
|
Ohm TG. Alterations of signal transduction in the lesioned entorhinal—hippocampal system: A mini‐review on alzheimer's disease‐related changes and experimental data. Hippocampus 2013. [DOI: 10.1002/hipo.1993.4500030716] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Thomas G. Ohm
- Zentrum der Morphologie, J.W. Goethe‐Universität, Frankfurt am Main, Germany
| |
Collapse
|
30
|
Sonntag WE, Deak F, Ashpole N, Toth P, Csiszar A, Freeman W, Ungvari Z. Insulin-like growth factor-1 in CNS and cerebrovascular aging. Front Aging Neurosci 2013; 5:27. [PMID: 23847531 PMCID: PMC3698444 DOI: 10.3389/fnagi.2013.00027] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/14/2013] [Indexed: 12/26/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is an important anabolic hormone that decreases with age. In the past two decades, extensive research has determined that the reduction in IGF-1 is an important component of the age-related decline in cognitive function in multiple species including humans. Deficiency in circulating IGF-1 results in impairment in processing speed and deficiencies in both spatial and working memory. Replacement of IGF-1 or factors that increase IGF-1 to old animals and humans reverses many of these cognitive deficits. Despite the overwhelming evidence for IGF-1 as an important neurotrophic agent, the specific mechanisms through which IGF-1 acts have remained elusive. Recent evidence indicates that IGF-1 is both produced by and has important actions on the cerebrovasculature as well as neurons and glia. Nevertheless, the specific regulation and actions of brain- and vascular-derived IGF-1 is poorly understood. The diverse effects of IGF-1 discovered thus far reveal a complex endocrine and paracrine system essential for integrating many of the functions necessary for brain health. Identification of the mechanisms of IGF-1 actions will undoubtedly provide critical insight into regulation of brain function in general and the causes of cognitive decline with age.
Collapse
Affiliation(s)
- William E Sonntag
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center Oklahoma City, OK, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Merino-Serrais P, Benavides-Piccione R, Blazquez-Llorca L, Kastanauskaite A, Rábano A, Avila J, DeFelipe J. The influence of phospho-τ on dendritic spines of cortical pyramidal neurons in patients with Alzheimer's disease. ACTA ACUST UNITED AC 2013; 136:1913-28. [PMID: 23715095 PMCID: PMC3673457 DOI: 10.1093/brain/awt088] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The dendritic spines on pyramidal cells represent the main postsynaptic elements of cortical excitatory synapses and they are fundamental structures in memory, learning and cognition. In the present study, we used intracellular injections of Lucifer yellow in fixed tissue to analyse over 19 500 dendritic spines that were completely reconstructed in three dimensions along the length of the basal dendrites of pyramidal neurons in the parahippocampal cortex and CA1 of patients with Alzheimer’s disease. Following intracellular injection, sections were immunostained for anti-Lucifer yellow and with tau monoclonal antibodies AT8 and PHF-1, which recognize tau phosphorylated at Ser202/Thr205 and at Ser396/404, respectively. We observed that the diffuse accumulation of phospho-tau in a putative pre-tangle state did not induce changes in the dendrites of pyramidal neurons, whereas the presence of tau aggregates forming intraneuronal neurofibrillary tangles was associated with progressive alteration of dendritic spines (loss of dendritic spines and changes in their morphology) and dendrite atrophy, depending on the degree of tangle development. Thus, the presence of phospho-tau in neurons does not necessarily mean that they suffer severe and irreversible effects as thought previously but rather, the characteristic cognitive impairment in Alzheimer’s disease is likely to depend on the relative number of neurons that have well developed tangles.
Collapse
Affiliation(s)
- Paula Merino-Serrais
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus Montegancedo S/N, 28223 Pozuelo de Alarcón, Spain
| | | | | | | | | | | | | |
Collapse
|
32
|
Amyloid plaque formation precedes dendritic spine loss. Acta Neuropathol 2012; 124:797-807. [PMID: 22993126 PMCID: PMC3508278 DOI: 10.1007/s00401-012-1047-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 09/07/2012] [Accepted: 09/09/2012] [Indexed: 11/01/2022]
Abstract
Amyloid-beta plaque deposition represents a major neuropathological hallmark of Alzheimer's disease. While numerous studies have described dendritic spine loss in proximity to plaques, much less is known about the kinetics of these processes. In particular, the question as to whether synapse loss precedes or follows plaque formation remains unanswered. To address this question, and to learn more about the underlying kinetics, we simultaneously imaged amyloid plaque deposition and dendritic spine loss by applying two-photon in vivo microscopy through a cranial window in double transgenic APPPS1 mice. As a result, we first observed that the rate of dendritic spine loss in proximity to plaques is the same in both young and aged animals. However, plaque size only increased significantly in the young cohort, indicating that spine loss persists even many months after initial plaque appearance. Tracking the fate of individual spines revealed that net spine loss is caused by increased spine elimination, with the rate of spine formation remaining constant. Imaging of dendritic spines before and during plaque formation demonstrated that spine loss around plaques commences at least 4 weeks after initial plaque formation. In conclusion, spine loss occurs, shortly but with a significant time delay, after the birth of new plaques, and persists in the vicinity of amyloid plaques over many months. These findings hence give further hope to the possibility that there is a therapeutic window between initial amyloid plaque deposition and the onset of structural damage at spines.
Collapse
|
33
|
The synaptic proteome in Alzheimer's disease. Alzheimers Dement 2012; 9:499-511. [PMID: 23154051 DOI: 10.1016/j.jalz.2012.04.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 12/28/2011] [Accepted: 04/25/2012] [Indexed: 11/23/2022]
Abstract
BACKGROUND Synaptic dysfunction occurs early in Alzheimer's disease (AD) and is recognized to be a primary pathological target for treatment. Synapse degeneration or dysfunction contributes to clinical signs of dementia through altered neuronal communication; the degree of synaptic loss correlates strongly with cognitive impairment. The molecular mechanisms underlying synaptic degeneration are still unclear, and identifying abnormally expressed synaptic proteins in AD brain will help to elucidate such mechanisms and to identify therapeutic targets that might slow AD progression. METHODS Synaptosomal fractions from human autopsy brain tissue from subjects with AD (n = 6) and without AD (n = 6) were compared using two-dimensional differential in-gel electrophoresis. AD pathology is region specific; human subjects can be highly variable in age, medication, and other factors. To counter these factors, two vulnerable areas (the hippocampus and the temporal cortex) were compared with two relatively spared areas (the motor and occipital cortices) within each group. Proteins exhibiting significant changes in expression were identified (≥20% change, Newman-Keuls P value < .05) using either matrix-assisted laser desorption ionization time-of-flight or electrospray ionisation quadrupole-time of flight mass spectrometry. RESULTS Twenty-six different synaptic proteins exhibited more than twofold differences in expression between AD and normal subjects. These proteins are involved in regulating different cellular functions, including energy metabolism, signal transduction, vesicle transport, structure, and antioxidant activity. CONCLUSION Comparative proteome analysis uncovered markers of pathogenic mechanisms involved in synaptic dysfunction.
Collapse
|
34
|
Zhou L, Hou Y, Yang Q, Du X, Li M, Yuan M, Zhou Z. Tetrahydroxystilbene glucoside improves the learning and memory of amyloid-β(₁₋₄₂)-injected rats and may be connected to synaptic changes in the hippocampus. Can J Physiol Pharmacol 2012. [PMID: 23181273 DOI: 10.1139/y2012-121] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this study was to evaluate the protective effects of 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucoside (TSG), an active component extracted from Polygonum multiflorum, on learning/memory deficits in Alzheimer's disease (AD). We randomly divided 24 male Sprague-Dawley rats among 4 groups: (i) the sham-operated group (control); (ii) sham-operated group also treated with TSG (sham+TSG); (iii) beta amyloid treated group (Aβ); and (iv) Aβ treatment group also treated with TSG (Aβ+TSG). Rats in the Aβ and Aβ+TSG groups were treated with Aβ₁₋₄₂ intracerebroventricularly, whereas the control and sham+TSG groups were given phosphate-buffered saline. Rats in the sham+TSG and Aβ+TSG groups were then treated intragastrically with TSG (50 mg·(kg body mass)⁻¹·day⁻¹) for 4 weeks, and rats in the Aβ and control groups were treated with saline. The results from Morris water maze tests, electron microscopy, real-time polymerase chain reaction, and Western blotting demonstrated that Aβ₁₋₄₂ induced impairment in learning and memory, degeneration in synaptic structures, and downregulation of Src and NR2B at the gene and protein level, respectively. These alterations were reversed by the administration of TSG, suggesting that TSG exerts anti-AD properties by protecting synaptic structure and function. TSG-induced upregulation of Src and NR2B may be responsible for this process.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | | | | | | | | | | | | |
Collapse
|
35
|
Bennett DA, Schneider JA, Arvanitakis Z, Wilson RS. Overview and findings from the religious orders study. Curr Alzheimer Res 2012; 9:628-45. [PMID: 22471860 PMCID: PMC3409291 DOI: 10.2174/156720512801322573] [Citation(s) in RCA: 498] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 12/27/2011] [Accepted: 01/12/2012] [Indexed: 12/22/2022]
Abstract
UNLABELLED The Religious Orders Study is a longitudinal clinical-pathologic cohort study of aging and Alzheimer's disease (AD). In this manuscript, we summarize the study methods including the study design and describe the clinical evaluation, assessment of risk factors, collection of ante-mortem biological specimens, brain autopsy and collection of selected postmortem data. THE RESULTS (1) review the relation of neuropathologic indices to clinical diagnoses and cognition proximate to death; (2) examine the relation of risk factors to clinical outcomes; (3) examine the relation of risk factors to measures of neuropathology; and (4) summarize additional study findings. We then discuss and contextualize the study findings.
Collapse
Affiliation(s)
- David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 600 S. Paulina, Suite 1028, Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
36
|
Deak F, Sonntag WE. Aging, synaptic dysfunction, and insulin-like growth factor (IGF)-1. J Gerontol A Biol Sci Med Sci 2012; 67:611-25. [PMID: 22503992 PMCID: PMC3348499 DOI: 10.1093/gerona/gls118] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 03/16/2012] [Indexed: 01/05/2023] Open
Abstract
Insulin-like growth factor (IGF)-1 is an important neurotrophic hormone. Deficiency of this hormone has been reported to influence the genesis of cognitive impairment and dementia in the elderly patients. Nevertheless, there are studies indicating that cognitive function can be maintained into old age even in the absence of circulating IGF-1 and studies that link IGF-1 to an acceleration of neurological diseases. Although IGF-1 has a complex role in brain function, synaptic effects appear to be central to the IGF-1-induced improvement in learning and memory. In this review, synaptic mechanisms of learning and memory and the effects of IGF-1 on synaptic communication are discussed. The emerging data indicate that synaptic function decreases with age and that IGF-1 contributes to information processing in the brain. Further studies that detail the specific actions of this important neurotrophic hormone will likely lead to therapies that result in improved cognitive function for the elderly patients.
Collapse
Affiliation(s)
- Ferenc Deak
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - William E. Sonntag
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, Oklahoma City, Oklahoma
| |
Collapse
|
37
|
Alves L, Correia ASA, Miguel R, Alegria P, Bugalho P. Alzheimer's disease: a clinical practice-oriented review. Front Neurol 2012; 3:63. [PMID: 22529838 PMCID: PMC3330267 DOI: 10.3389/fneur.2012.00063] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 04/02/2012] [Indexed: 12/12/2022] Open
Abstract
Investigation in the field of Alzheimer's disease (AD), the commonest cause of dementia, has been very active in recent years and it may be difficult for the clinician to keep up with all the innovations and to be aware of the implications they have in clinical practice. The authors, thus, reviewed recent literature on the theme in order to provide the clinician with an updated overview, intended to support decision-making on aspects of diagnosis and management. This article begins to focus on the concept of AD and on its pathogenesis. Afterward, epidemiology and non-genetic risk factors are approached. Genetics, including genetic risk factors and guidelines for genetic testing, are mentioned next. Recommendations for diagnosis of AD, including recently proposed criteria, are then reviewed. Data on the variants of AD is presented. First approach to the patient is dealt with next, followed by neuropsychological evaluation. Biomarkers, namely magnetic resonance imaging, single photon emission tomography, FDG PET, PiB PET, CSF tau, and Aβ analysis, as well as available data on their diagnostic accuracy, are also discussed. Factors predicting rate of disease progression are briefly mentioned. Finally, non-pharmacological and pharmacological treatments, including established and emerging drugs, are addressed.
Collapse
Affiliation(s)
- Luísa Alves
- Serviço de Neurologia, Hospital de Egas Moniz, Centro Hospitalar de Lisboa OcidentalLisboa, Portugal
- Faculdade de Ciências Médicas, Universidade nova de LisboaLisboa, Portugal
- Centro de Estudos de Doenças CrónicasLisboa, Portugal
| | - Ana Sofia A. Correia
- Serviço de Neurologia, Hospital de Egas Moniz, Centro Hospitalar de Lisboa OcidentalLisboa, Portugal
| | - Rita Miguel
- Serviço de Neurologia, Hospital de Egas Moniz, Centro Hospitalar de Lisboa OcidentalLisboa, Portugal
| | | | - Paulo Bugalho
- Serviço de Neurologia, Hospital de Egas Moniz, Centro Hospitalar de Lisboa OcidentalLisboa, Portugal
- Faculdade de Ciências Médicas, Universidade nova de LisboaLisboa, Portugal
- Centro de Estudos de Doenças CrónicasLisboa, Portugal
| |
Collapse
|
38
|
Counts SE, He B, Nadeem M, Wuu J, Scheff SW, Mufson EJ. Hippocampal drebrin loss in mild cognitive impairment. NEURODEGENER DIS 2012; 10:216-9. [PMID: 22310934 DOI: 10.1159/000333122] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 09/03/2011] [Indexed: 11/19/2022] Open
Abstract
Alterations in the relative abundance of synaptic proteins may contribute to hippocampal synaptic dysfunction in Alzheimer's disease (AD). The extent to which perturbations in synaptic protein expression occur during the earliest stages of cognitive decline remains unclear. We examined protein levels of presynaptic synaptophysin (SYP) and synaptotagmin (SYT), and postsynaptic drebrin (DRB), a marker for dendritic spine plasticity, in the hippocampus of people with an antemortem clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI) or mild/moderate AD. Although normalized SYP and SYT levels were preserved, DRB was reduced by approximately 40% in the hippocampus of MCI and AD compared to NCI subjects. This differential alteration of synaptic markers in MCI suggests a selective impairment in hippocampal postsynaptic dendritic plasticity in prodromal AD that likely heralds the onset of memory impairment in symptomatic disease.
Collapse
Affiliation(s)
- Scott E Counts
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Ill 60612, USA
| | | | | | | | | | | |
Collapse
|
39
|
Synaptic dysfunction in Alzheimer's disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 970:573-601. [PMID: 22351073 DOI: 10.1007/978-3-7091-0932-8_25] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Generation of amyloid peptide (Aβ) is at the beginning of a cascade that leads to Alzheimer's disease (AD). Amyloid precursor protein (APP), as well as β- and γ-secretases, is the principal player involved in Aβ production, while α-secretase cleavage on APP prevents Aβ deposition. Recent studies suggested that soluble assembly states of Aβ peptides can cause cognitive problems by disrupting synaptic function in the absence of significant neurodegeneration. Therefore, current research investigates the relative importance of these various soluble Aβ assemblies in causing synaptic dysfunction and cognitive deficits. Several Aβ oligomers targets and cellular mechanisms responsible of Aβ-induced synaptic failure have been identified. The first and most important mechanism impugns a toxic gain of function for Aβ which results due to self-association and attainment of new structures capable of novel interactions that lead to impaired plasticity. Other scenarios predicate that Aβ has a normal physiological role. On the one hand, insufficient Aβ could lead to a loss of normal function, whereas excess Aβ may precipitate dysfunction. How this occurs and which the main target/s is/are for the synaptic action of Aβ remains to be fully understood and would certainly represent one of the main challenges to future AD research.
Collapse
|
40
|
Changes in hippocampal synapses and learning-memory abilities in a streptozotocin-treated rat model and intervention by using fasudil hydrochloride. Neuroscience 2012; 200:120-9. [DOI: 10.1016/j.neuroscience.2011.10.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 10/15/2011] [Accepted: 10/17/2011] [Indexed: 01/03/2023]
|
41
|
Mufson EJ, Binder L, Counts SE, DeKosky ST, de Toledo-Morrell L, Ginsberg SD, Ikonomovic MD, Perez SE, Scheff SW. Mild cognitive impairment: pathology and mechanisms. Acta Neuropathol 2012; 123:13-30. [PMID: 22101321 PMCID: PMC3282485 DOI: 10.1007/s00401-011-0884-1] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 09/29/2011] [Accepted: 09/30/2011] [Indexed: 10/15/2022]
Abstract
Mild cognitive impairment (MCI) is rapidly becoming one of the most common clinical manifestations affecting the elderly. The pathologic and molecular substrate of people diagnosed with MCI is not well established. Since MCI is a human specific disorder and neither the clinical nor the neuropathological course appears to follow a direct linear path, it is imperative to characterize neuropathology changes in the brains of people who came to autopsy with a well-characterized clinical diagnosis of MCI. Herein, we discuss findings derived from clinical pathologic studies of autopsy cases who died with a clinical diagnosis of MCI. The heterogeneity of clinical MCI imparts significant challenges to any review of this subject. The pathologic substrate of MCI is equally complex and must take into account not only conventional plaque and tangle pathology but also a wide range of cellular, biochemical and molecular deficits, many of which relate to cognitive decline as well as compensatory responses to the progressive disease process. The multifaceted nature of the neuronal disconnection syndrome associated with MCI suggests that there is no single event which precipitates this prodromal stage of AD. In fact, it can be argued that neuronal degeneration initiated at different levels of the central nervous system drives cognitive decline as a final common pathway at this stage of the dementing disease process.
Collapse
Affiliation(s)
- Elliott J Mufson
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St., Suite 300, Chicago, IL 60612, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wang X, Michaelis ML, Michaelis EK. Functional genomics of brain aging and Alzheimer's disease: focus on selective neuronal vulnerability. Curr Genomics 2011; 11:618-33. [PMID: 21629439 PMCID: PMC3078686 DOI: 10.2174/138920210793360943] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/05/2010] [Accepted: 10/21/2010] [Indexed: 01/02/2023] Open
Abstract
Pivotal brain functions, such as neurotransmission, cognition, and memory, decline with advancing age and, especially, in neurodegenerative conditions associated with aging, such as Alzheimer’s disease (AD). Yet, deterioration in structure and function of the nervous system during aging or in AD is not uniform throughout the brain. Selective neuronal vulnerability (SNV) is a general but sometimes overlooked characteristic of brain aging and AD. There is little known at the molecular level to account for the phenomenon of SNV. Functional genomic analyses, through unbiased whole genome expression studies, could lead to new insights into a complex process such as SNV. Genomic data generated using both human brain tissue and brains from animal models of aging and AD were analyzed in this review. Convergent trends that have emerged from these data sets were considered in identifying possible molecular and cellular pathways involved in SNV. It appears that during normal brain aging and in AD, neurons vulnerable to injury or cell death are characterized by significant decreases in the expression of genes related to mitochondrial metabolism and energy production. In AD, vulnerable neurons also exhibit down-regulation of genes related to synaptic neurotransmission and vesicular transport, cytoskeletal structure and function, and neurotrophic factor activity. A prominent category of genes that are up-regulated in AD are those related to inflammatory response and some components of calcium signaling. These genomic differences between sensitive and resistant neurons can now be used to explore the molecular underpinnings of previously suggested mechanisms of cell injury in aging and AD.
Collapse
Affiliation(s)
- Xinkun Wang
- Higuchi Biosciences Center and Department of Pharmacology and Toxicology, The University of Kansas, Lawrence, KS 66047, USA
| | | | | |
Collapse
|
43
|
Krafft GA, Klein WL. ADDLs and the signaling web that leads to Alzheimer’s disease. Neuropharmacology 2010; 59:230-42. [DOI: 10.1016/j.neuropharm.2010.07.012] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 07/13/2010] [Indexed: 12/29/2022]
|
44
|
Podoly E, Hanin G, Soreq H. Alanine-to-threonine substitutions and amyloid diseases: butyrylcholinesterase as a case study. Chem Biol Interact 2010; 187:64-71. [PMID: 20060816 DOI: 10.1016/j.cbi.2010.01.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Revised: 12/23/2009] [Accepted: 01/04/2010] [Indexed: 01/04/2023]
Abstract
Alanine-to-threonine (A to T) substitutions caused by single nucleotide polymorphisms (SNPs) occur in diverse proteins, and in certain cases these substitutions induce self-aggregation into amyloid fibrils or aggregation in other amyloidogenic proteins. This is compatible with the inverse preferences of alanine to form helices and of threonine to support beta-sheet structures, which are crucial for amyloid fibrils formation. Our interest in these mutations was initiated by studying the potential effects of the A539T substitution in the butyrylcholinesterase BChE-K variant on amyloid fibrils formation in Alzheimer's disease. Other examples are, Parkinson's disease (PD), where A53T alpha-synuclein occurs in Lewy bodies and familial amyloid polyneuropathy (FAP), where an A25T substitution appears in transthyretin (TTR). In peripheral organs, an A34T substitution is found in the light chain immunoglobulin genes of patients with systemic amyloidosis and in familial hypercholesterolemia, an A370T substitution occurs in the LDLR regulator of cholesterol homeostasis. That such substitutions appear in proteins with important cellular functions suggests that they confer antagonistic pleiotropy, providing added value at an earlier age but causing damages and inducing amyloid diseases later on. This, in turn, may explain the evolutionary selection and preservation of these substitutions. The structural effect of residue substitutions and in particular A to T substitutions in amyloidogenic diseases thus merits further attention.
Collapse
Affiliation(s)
- Erez Podoly
- Department of Biological Chemistry, The Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | | | | |
Collapse
|
45
|
Robertson RT, Baratta J, Yu J, LaFerla FM. Amyloid-beta expression in retrosplenial cortex of triple transgenic mice: relationship to cholinergic axonal afferents from medial septum. Neuroscience 2009; 164:1334-46. [PMID: 19772895 PMCID: PMC2784206 DOI: 10.1016/j.neuroscience.2009.09.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 09/08/2009] [Accepted: 09/14/2009] [Indexed: 12/17/2022]
Abstract
Triple transgenic (3xTg-AD) mice harboring the presenilin 1, amyloid precursor protein, and tau transgenes (Oddo et al., 2003b) display prominent levels of amyloid-beta (Abeta) immunoreactivity in forebrain regions. The Abeta immunoreactivity is first seen intracellularly in neurons and later as extracellular plaque deposits. The present study examined Abeta immunoreactivity that occurs in layer III of the granular division of retrosplenial cortex (RSg). This pattern of Abeta immunoreactivity in layer III of RSg develops relatively late, and is seen in animals older than 14 months. The appearance of the Abeta immunoreactivity is similar to an axonal terminal field and thus may offer a unique opportunity to study the relationship between afferent projections and the formation of Abeta deposits. Axonal tract tracing techniques demonstrated that the pattern of axon terminal labeling in layer III of RSg, following placement of DiI in medial septum, is remarkably similar to the pattern of cholinergic axons in RSg, as detected by acetylcholinesterase histochemical staining, choline acetyltransferase immunoreactivity, or p75 receptor immunoreactivity; this pattern also is strikingly similar to the band of Abeta immunoreactivity. In animals sustaining early damage to the medial septal nucleus (prior to the advent of Abeta immunoreactivity), the band of Abeta in layer III of RSg does not develop; the corresponding band of cholinergic markers also is eliminated. In older animals (after the appearance of the Abeta immunoreactivity) damage to cholinergic afferents by electrolytic lesions, immunotoxin lesions, or cutting the cingulate bundle, result in a rapid loss of the cholinergic markers and a slower reduction of Abeta immunoreactivity. These results suggest that the septal cholinergic axonal projections transport Abeta or amyloid precursor protein (APP) to layer III of RSg.
Collapse
Affiliation(s)
- R T Robertson
- Department of Anatomy and Neurobiology, University of California, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
46
|
Arendt T. Synaptic degeneration in Alzheimer's disease. Acta Neuropathol 2009; 118:167-79. [PMID: 19390859 DOI: 10.1007/s00401-009-0536-x] [Citation(s) in RCA: 373] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 04/07/2009] [Accepted: 04/07/2009] [Indexed: 01/03/2023]
Abstract
Synaptic loss is the major neurobiological substrate of cognitive dysfunction in Alzheimer's disease (AD). Synaptic failure is an early event in the pathogenesis that is clearly detectable already in patients with mild cognitive impairment (MCI), a prodromal state of AD. It progresses during the course of AD and in most early stages involves mechanisms of compensation before reaching a stage of decompensated function. This dynamic process from an initially reversible functionally responsive stage of down-regulation of synaptic function to stages irreversibly associated with degeneration might be related to a disturbance of structural brain self-organization and involves morpho-regulatory molecules such as the amyloid precursor protein. Further, recent evidence suggests a role for diffusible oligomers of amyloid beta in synaptic dysfunction. To form synaptic connections and to continuously re-shape them in a process of ongoing structural adaptation, neurons must permanently withdraw from the cell cycle. Previously, we formulated the hypothesis that differentiated neurons after having withdrawn from the cell cycle are able to use molecular mechanisms primarily developed to control proliferation alternatively to control synaptic plasticity. The existence of these alternative effector pathways within neurons might put them at risk of erroneously converting signals derived from plastic synaptic changes into the program of cell cycle activation, which subsequently leads to cell death. The molecular mechanisms involved in cell cycle activation might, thus, link aberrant synaptic changes to cell death.
Collapse
Affiliation(s)
- Thomas Arendt
- Paul Flechsig Institute of Brain Research, University of Leipzig, Jahnallee 59, 04109 Leipzig, Germany.
| |
Collapse
|
47
|
Head E, Corrada MM, Kahle-Wrobleski K, Kim RC, Sarsoza F, Goodus M, Kawas CH. Synaptic proteins, neuropathology and cognitive status in the oldest-old. Neurobiol Aging 2007; 30:1125-34. [PMID: 18006193 DOI: 10.1016/j.neurobiolaging.2007.10.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 09/28/2007] [Accepted: 10/04/2007] [Indexed: 11/16/2022]
Abstract
An increasing number of individuals in our population are surviving to over 90 years and a subset is at risk for developing dementia. However, senile plaque and neurofibrillary tangle pathology do not consistently differentiate individuals with and without dementia. Synaptic protein loss is a feature of aging and dementia and may dissociate 90+ individuals with and without dementia. Synaptophysin (SYN), postsynaptic density 95 (PSD-95) and growth-associated protein 43 (GAP-43) were studied in the frontal cortex of an autopsy series of 32 prospectively followed individuals (92-105 years) with a range of cognitive function. SYN protein levels were decreased in individuals with dementia and increased in those with clinical signs of cognitive impairment insufficient for a diagnosis of dementia. SYN but neither PSD-95 nor GAP-43 protein levels were significantly correlated with mini-mental status examination (MMSE) scores. Frontal cortex SYN protein levels may protect neuronal function in oldest-old individuals and reflect compensatory responses that may be involved with maintaining cognition.
Collapse
Affiliation(s)
- Elizabeth Head
- Institute of Brain Aging and Dementia, University of California, Irvine, CA 92697-4540, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Ryzhikov S, Bahr BA. Gephyrin alterations due to protein accumulation stress are reduced by the lysosomal modulator Z-Phe-Ala-diazomethylketone. J Mol Neurosci 2007; 34:131-9. [PMID: 18204977 DOI: 10.1007/s12031-007-9009-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2007] [Accepted: 08/22/2007] [Indexed: 11/25/2022]
Abstract
Inhibitory neurotransmission is important for brain function and requires specific transmitter receptors that are organized in synaptic domains. Gephyrin is a cytoskeletal organization protein that binds tubulin and plays an important role in clustering and organizing select inhibitory neurotransmitter receptors. Here, we tested if gephyrin is altered by protein accumulation stress that is common in age-related neurodegenerative disorders. For this, we used the hippocampal slice model that has been shown to exhibit chloroquine (CQN)-induced protein accumulation, microtubule destabilization, transport failure, and declines in excitatory neurotransmitter receptors and their responses. In addition to the decreases in excitatory receptor subunits and other glutamatergic markers, we found that gephyrin isoforms were reduced across the CQN treatment period. Associated with this decline in gephyrin levels was the production of three gephyrin breakdown products (GBDPs) of 30, 38, and 48 kDa. The induced effects on gephyrin were tested for evidence of recovery through enhancement of lysosomal function that is known to promote protein clearance and microtubule integrity. Using the lysosomal modulator Z-Phe-Ala-diazomethylketone (PADK), gephyrin levels were completely restored in correspondence with the recovery of excitatory glutamatergic components. In addition, GBDPs were significantly reduced after the 2-day PADK treatment, to levels that were at or below those measured in control cultures. These findings suggest that receptor-clustering mechanisms for inhibitory synapses are compromised during protein accumulation events. They also indicate that a lysosomal enhancement strategy can protect gephyrin integrity, which may be vital for the balance between inhibitory and excitatory signaling during age-related diseases.
Collapse
Affiliation(s)
- Sophia Ryzhikov
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | | |
Collapse
|
49
|
Butler D, Bendiske J, Michaelis ML, Karanian DA, Bahr BA. Microtubule-stabilizing agent prevents protein accumulation-induced loss of synaptic markers. Eur J Pharmacol 2007; 562:20-7. [PMID: 17336290 DOI: 10.1016/j.ejphar.2007.01.053] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Revised: 01/15/2007] [Accepted: 01/17/2007] [Indexed: 11/18/2022]
Abstract
Synaptic pathology is associated with protein accumulation events, and is thought by many to be the best correlate of cognitive impairment in normal aging and different types of dementia including Alzheimer's disease. Numerous studies point to the disruption of microtubule-based transport mechanisms as a contributor to synaptic degeneration. Reported reductions in a microtubule stability marker, acetylated alpha-tubulin, suggest that disrupted transport occurs in Alzheimer's disease neurons, and such a reduction is known to be associated with transport failure and synaptic compromise in a hippocampal slice model of protein accumulation. The slice model exhibits accumulated proteins in response to chloroquine-mediated lysosomal dysfunction, resulting in corresponding decreases in acetylated tubulin and pre- and postsynaptic markers (synaptophysin and glutamate receptors). To test whether the protein deposition-induced loss of synaptic proteins is due to disruption of microtubule integrity, a potent microtubule-stabilizing agent, the taxol derivative TX67 (10-succinyl paclitaxel), was applied to the hippocampal slice cultures. In the absence of lysosomal stress, TX67 (100-300 nM) provided microtubule stabilization as indicated by markedly increased levels of acetylated tubulin. When TX67 was applied to the slices during the chloroquine treatment period, pre- and postsynaptic markers were maintained at control levels. In addition, a correlation was evident across slice samples between levels of acetylated tubulin and glutamate receptor subunit GluR1. These data indicate that disruption of microtubule integrity accounts for protein deposition-induced synaptic decline. They also suggest that microtubule-stabilizing drugs can be used to slow or halt the progressive synaptic deterioration linked to Alzheimer-type pathogenesis.
Collapse
Affiliation(s)
- David Butler
- Department of Pharmaceutical Sciences and the Neurosciences Program, University of Connecticut, Storrs, CT 06269-3092, USA
| | | | | | | | | |
Collapse
|
50
|
Katsuse O, Lin WL, Lewis J, Hutton ML, Dickson DW. Neurofibrillary tangle-related synaptic alterations of spinal motor neurons of P301L tau transgenic mice. Neurosci Lett 2006; 409:95-9. [PMID: 17010516 DOI: 10.1016/j.neulet.2006.09.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2006] [Revised: 09/08/2006] [Accepted: 09/09/2006] [Indexed: 11/30/2022]
Abstract
We investigated axosomatic synapses of anterior horn cells of transgenic (TG) mice expressing mutant P301L human tau and non-transgenic (NTG) mice using electron microscopic methods to demonstrate the relationship between neurofibrillary tangles (NFTs) and synaptic alterations. Animals aged 3.5-8.5 months were used because at this age many motor neurons in TG mice have NFTs. We measured the perimeter of anterior horn cell perikarya, the number of boutons and total length of boutons in contact with the neuronal perikarya from the micrographs of NFT and non-NFT-bearing neurons. We also calculated the proportion of the perimeter covered by boutons, density of boutons and mean size of boutons. The density of synaptic boutons in contact with NFT-bearing neurons was significantly decreased compared to non-NFT-bearing neurons. These findings suggest that synaptic reduction occurs during neurofibrillary degeneration and is probably associated with NFT. In addition, synaptic boutons were detached from NFT-bearing neurons with the resulting space occupied by astrocytic processes, suggesting that astrocytes may be involved in the observed synaptic alterations.
Collapse
Affiliation(s)
- Omi Katsuse
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | | | | | | | | |
Collapse
|