51
|
Pilozzi A, Yu Z, Carreras I, Cormier K, Hartley D, Rogers J, Dedeoglu A, Huang X. A Preliminary Study of Cu Exposure Effects upon Alzheimer's Amyloid Pathology. Biomolecules 2020; 10:E408. [PMID: 32155778 PMCID: PMC7175127 DOI: 10.3390/biom10030408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 01/25/2023] Open
Abstract
A large body of evidence indicates that dysregulation of cerebral biometals (Fe, Cu, Zn) and their interactions with amyloid precursor protein (APP) and Aβ amyloid may contribute to the Alzheimer's disease (AD) Aβ amyloid pathology. However, the molecular underpinnings associated with the interactions are still not fully understood. Herein we have further validated the exacerbation of Aβ oligomerization by Cu and H2O2 in vitro. We have also reported that Cu enhanced APP translations via its 5' untranslated region (5'UTR) of mRNA in SH-SY5Y cells, and increased Aβ amyloidosis and expression of associated pro-inflammatory cytokines such as MCP-5 in Alzheimer's APP/PS1 doubly transgenic mice. This preliminary study may further unravel the pathogenic role of Cu in Alzheimer's Aβ amyloid pathogenesis, warranting further investigation.
Collapse
Affiliation(s)
- Alexander Pilozzi
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA; (A.P.); (J.R.)
| | - Zhanyang Yu
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA;
| | - Isabel Carreras
- Department of Veterans Affairs, VA Medical Center, Bedford, MA 01730, USA; (I.C.); (K.C.); (A.D.)
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kerry Cormier
- Department of Veterans Affairs, VA Medical Center, Bedford, MA 01730, USA; (I.C.); (K.C.); (A.D.)
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | | | - Jack Rogers
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA; (A.P.); (J.R.)
| | - Alpaslan Dedeoglu
- Department of Veterans Affairs, VA Medical Center, Bedford, MA 01730, USA; (I.C.); (K.C.); (A.D.)
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA; (A.P.); (J.R.)
| |
Collapse
|
52
|
Zhang X, Zhao F, Wang C, Zhang J, Bai Y, Zhou F, Wang Z, Wu M, Yang W, Guo J, Qi J. AVP(4-8) Improves Cognitive Behaviors and Hippocampal Synaptic Plasticity in the APP/PS1 Mouse Model of Alzheimer's Disease. Neurosci Bull 2019; 36:254-262. [PMID: 31605298 DOI: 10.1007/s12264-019-00434-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 07/12/2019] [Indexed: 11/26/2022] Open
Abstract
Memory deficits with aging are related to the neurodegeneration in the brain, including a reduction in arginine vasopressin (AVP) in the brain of patients with Alzheimer's disease (AD). AVP(4-8), different from its precursor AVP, plays memory enhancement roles in the CNS without peripheral side-effects. However, it is not clear whether AVP(4-8) can improve cognitive behaviors and synaptic plasticity in the APP/PS1 mouse model of AD. Here, we investigated for the first time the neuroprotective effects of AVP(4-8) on memory behaviors and in vivo long-term potentiation (LTP) in APP/PS1-AD mice. The results showed that: (1) APP/PS1-AD mice had lower spontaneous alternation in the Y-maze than wild-type (WT) mice, and this was significantly reversed by AVP(4-8); (2) the prolonged escape latency of APP/PS1-AD mice in the Morris water maze was significantly decreased by AVP(4-8), and the decreased swimming time in target quadrant recovered significantly after AVP(4-8) treatment; (3) in vivo hippocampal LTP induced by high-frequency stimulation had a significant deficit in the AD mice, and this was partly rescued by AVP(4-8); (4) AVP(4-8) significantly up-regulated the expression levels of postsynaptic density 95 (PSD95) and nerve growth factor (NGF) in the hippocampus of AD mice. These results reveal the beneficial effects of AVP(4-8) in APP/PS1-AD mice, showing that the intranasal administration of AVP(4-8) effectively improved the working memory and long-term spatial memory of APP/PS1-AD mice, which may be associated with the elevation of PSD95 and NGF levels in the brain and the maintenance of hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Xiumin Zhang
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan, 030001, China
- Department of Physiology, Key laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Fang Zhao
- Department of Physiology, Key laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Chenfang Wang
- Department of Physiology, Key laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Jun Zhang
- Department of Physiology, Key laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Yu Bai
- Department of Physiology, Key laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Fang Zhou
- Department of Physiology, Key laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Zhaojun Wang
- Department of Physiology, Key laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Meina Wu
- Department of Physiology, Key laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Wei Yang
- Department of Physiology, Key laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Junhong Guo
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan, 030001, China.
| | - Jinshun Qi
- Department of Physiology, Key laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
53
|
Chen MK, Mecca AP, Naganawa M, Finnema SJ, Toyonaga T, Lin SF, Najafzadeh S, Ropchan J, Lu Y, McDonald JW, Michalak HR, Nabulsi NB, Arnsten AFT, Huang Y, Carson RE, van Dyck CH. Assessing Synaptic Density in Alzheimer Disease With Synaptic Vesicle Glycoprotein 2A Positron Emission Tomographic Imaging. JAMA Neurol 2019; 75:1215-1224. [PMID: 30014145 DOI: 10.1001/jamaneurol.2018.1836] [Citation(s) in RCA: 281] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance Synaptic loss is well established as the major structural correlate of cognitive impairment in Alzheimer disease (AD). The ability to measure synaptic density in vivo could accelerate the development of disease-modifying treatments for AD. Synaptic vesicle glycoprotein 2A is an essential vesicle membrane protein expressed in virtually all synapses and could serve as a suitable target for synaptic density. Objective To compare hippocampal synaptic vesicle glycoprotein 2A (SV2A) binding in participants with AD and cognitively normal participants using positron emission tomographic (PET) imaging. Design, Setting, and Participants This cross-sectional study recruited 10 participants with AD and 11 participants who were cognitively normal between November 2015 and June 2017. We hypothesized a reduction in hippocampal SV2A binding in AD, based on the early degeneration of entorhinal cortical cell projections to the hippocampus (via the perforant path) and hippocampal SV2A reductions that had been observed in postmortem studies. Participants underwent high-resolution PET scanning with ((R)-1-((3-(11C-methyl-11C)pyridin-4-yl)methyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one), a compound more commonly known as 11C-UCB-J, for SV2A. They also underwent high-resolution PET scanning with carbon 11-labeled Pittsburgh Compound B (11C-PiB) for β-amyloid, magnetic resonance imaging, and cognitive and neurologic evaluation. Main Outcomes and Measures Outcomes were 11C-UCB-J-specific binding (binding potential [BPND]) via PET imaging in brain regions of interest in participants with AD and participants who were cognitively normal. Results Ten participants with AD (5 male and 5 female; mean [SD] age, 72.7 [6.3] years; 10 [100%] β-amyloid positive) were compared with 11 participants who were cognitively normal (5 male and 6 female; mean [SD] age, 72.9 [8.7] years; 11 [100%] β-amyloid negative). Participants with AD spanned the disease stages from amnestic mild cognitive impairment (n = 5) to mild dementia (n = 5). Participants with AD had significant reduction in hippocampal SV2A specific binding (41%) compared with cognitively normal participants, as assessed by 11C-UCB-J-PET BPND (cognitively normal participants: mean [SD] BPND, 1.47 [0.37]; participants with AD: 0.87 [0.50]; P = .005). These reductions remained significant after correction for atrophy (ie, partial volume correction; participants who were cognitively normal: mean [SD], 2.71 [0.46]; participants with AD: 2.15 [0.55]; P = .02). Hippocampal SV2A-specific binding BPND was correlated with a composite episodic memory score in the overall sample (R = 0.56; P = .01). Conclusions and Relevance To our knowledge, this is the first study to investigate synaptic density in vivo in AD using 11C-UCB-J-PET imaging. This approach may provide a direct measure of synaptic density, and it therefore holds promise as an in vivo biomarker for AD and as an outcome measure for trials of disease-modifying therapies, particularly those targeted at the preservation and restoration of synapses.
Collapse
Affiliation(s)
- Ming-Kai Chen
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Adam P Mecca
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Sjoerd J Finnema
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Shu-Fei Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Soheila Najafzadeh
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Jim Ropchan
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Yihuan Lu
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Julia W McDonald
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Hannah R Michalak
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Amy F T Arnsten
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
54
|
AD molecular: Molecular imaging of Alzheimer's disease: PET imaging of neurotransmitter systems. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019. [PMID: 31481161 DOI: 10.1016/bs.pmbts.2019.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Current understanding of Alzheimer's disease (AD) pathogenesis relies on the observed accumulations of amyloid β and phosphorylated tau aggregates that are thought to play key roles in initiating or propagating disease. However, other processes including changes in synaptic proteins and neurotransmitter loss have been suggested as important etiologies or contributors. Positron emission tomography (PET) imaging allows in vivo investigations of molecular changes associated with AD. PET imaging with multiple radiotracers can be used in combination with other modalities such as magnetic resonance imaging (MRI), and with assessments of cognition and neuropsychiatric symptoms to investigate the molecular underpinnings of AD. Studies of synaptic protein changes may improve the understanding of disease mechanisms and provide valuable markers of disease progression and therapeutic efficacy. This chapter will illustrate the importance of in vivo molecular imaging in the study of AD with a specific emphasis on PET and radioligands for several non-amyloid targets.
Collapse
|
55
|
Lavado LK, Zhang MH, Patel K, Khan S, Patel UK. Biometals as Potential Predictors of the Neurodegenerative Decline in Alzheimer's Disease. Cureus 2019; 11:e5573. [PMID: 31695992 PMCID: PMC6820671 DOI: 10.7759/cureus.5573] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s Disease (AD) is a debilitating neurodegenerative disease that is diagnosed by gradual memory loss and certain cognitive impairments involving attention, reasoning, and language. Most of the research on Alzheimer’s disease focuses on the correlation of its neuropathological changes in the neurofibrillary tangles caused by hyper-phosphorylated tau protein and β-amyloid plaques with respect to cognitive impairment. Its pathology, however, remains incompletely understood. Currently, research has demonstrated that environmental factors such as biometals play a crucial role in exacerbating AD progression. The present review examines the role of metals in AD progression and how metal dyshomeostasis attributes to AD pathogenesis. It was found that certain metals possess both beneficial and harmful properties in terms of AD progression. Depending upon the concentration of the metal of interest, copper, zinc, iron, and selenium have general beneficial properties. However, when present in excess, they can lead to oxidative stress and hyperphosphorylation of tau protein, amongst other harmful effects, while calcium and magnesium were seen to have beneficial effects by regulating biometal uptake. In this review, we have provided evidential studies that focus on the involvement of certain metals in antioxidant pathways leading to the formation of reactive species indicative of neurodegeneration.
Collapse
Affiliation(s)
| | - Michelle H Zhang
- Psychological & Brain Sciences and Biology, Johns Hopkins University, Baltimore, USA
| | - Karan Patel
- Neuroscience, Johns Hopkins University, Baltimore, USA
| | - Sohim Khan
- Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Urvish K Patel
- Neurology and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
56
|
Bosiacki M, Gąssowska-Dobrowolska M, Kojder K, Fabiańska M, Jeżewski D, Gutowska I, Lubkowska A. Perineuronal Nets and Their Role in Synaptic Homeostasis. Int J Mol Sci 2019; 20:ijms20174108. [PMID: 31443560 PMCID: PMC6747153 DOI: 10.3390/ijms20174108] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
Extracellular matrix (ECM) molecules that are released by neurons and glial cells form perineuronal nets (PNNs) and modulate many neuronal and glial functions. PNNs, whose structure is still not known in detail, surround cell bodies and dendrites, which leaves free space for synapses to come into contact. A reduction in the expression of many neuronal ECM components adversely affects processes that are associated with synaptic plasticity, learning, and memory. At the same time, increased ECM activity, e.g., as a result of astrogliosis following brain damage or in neuroinflammation, can also have harmful consequences. The therapeutic use of enzymes to attenuate elevated neuronal ECM expression after injury or in Alzheimer’s disease has proven to be beneficial by promoting axon growth and increasing synaptic plasticity. Yet, severe impairment of ECM function can also lead to neurodegeneration. Thus, it appears that to ensure healthy neuronal function a delicate balance of ECM components must be maintained. In this paper we review the structure of PNNs and their components, such as hyaluronan, proteoglycans, core proteins, chondroitin sulphate proteoglycans, tenascins, and Hapln proteins. We also characterize the role of ECM in the functioning of the blood-brain barrier, neuronal communication, as well as the participation of PNNs in synaptic plasticity and some clinical aspects of perineuronal net impairment. Furthermore, we discuss the participation of PNNs in brain signaling. Understanding the molecular foundations of the ways that PNNs participate in brain signaling and synaptic plasticity, as well as how they change in physiological and pathological conditions, may help in the development of new therapies for many degenerative and inflammatory diseases of the brain.
Collapse
Affiliation(s)
- Mateusz Bosiacki
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University in Szczecin, Żołnierska 54 Str., 71-210 Szczecin, Poland
| | - Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 Str., 02-106 Warsaw, Poland
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Marta Fabiańska
- Institute of Philosophy, University of Szczecin, Krakowska 71-79 Str., 71-017 Szczecin, Poland
| | - Dariusz Jeżewski
- Department of Neurosurgery and Pediatric Neurosurgery, Department of Applied Neurocognitivistics, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Izabela Gutowska
- Department of Human Nutrition and Metabolomics, Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Broniewskiego 24 Str., 71-252 Szczecin, Poland
| | - Anna Lubkowska
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University in Szczecin, Żołnierska 54 Str., 71-210 Szczecin, Poland.
| |
Collapse
|
57
|
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: 184] [Impact Index Per Article: 36.8] [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
|
58
|
Cline EN, Bicca MA, Viola KL, Klein WL. The Amyloid-β Oligomer Hypothesis: Beginning of the Third Decade. J Alzheimers Dis 2019; 64:S567-S610. [PMID: 29843241 PMCID: PMC6004937 DOI: 10.3233/jad-179941] [Citation(s) in RCA: 543] [Impact Index Per Article: 108.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The amyloid-β oligomer (AβO) hypothesis was introduced in 1998. It proposed that the brain damage leading to Alzheimer’s disease (AD) was instigated by soluble, ligand-like AβOs. This hypothesis was based on the discovery that fibril-free synthetic preparations of AβOs were potent CNS neurotoxins that rapidly inhibited long-term potentiation and, with time, caused selective nerve cell death (Lambert et al., 1998). The mechanism was attributed to disrupted signaling involving the tyrosine-protein kinase Fyn, mediated by an unknown toxin receptor. Over 4,000 articles concerning AβOs have been published since then, including more than 400 reviews. AβOs have been shown to accumulate in an AD-dependent manner in human and animal model brain tissue and, experimentally, to impair learning and memory and instigate major facets of AD neuropathology, including tau pathology, synapse deterioration and loss, inflammation, and oxidative damage. As reviewed by Hayden and Teplow in 2013, the AβO hypothesis “has all but supplanted the amyloid cascade.” Despite the emerging understanding of the role played by AβOs in AD pathogenesis, AβOs have not yet received the clinical attention given to amyloid plaques, which have been at the core of major attempts at therapeutics and diagnostics but are no longer regarded as the most pathogenic form of Aβ. However, if the momentum of AβO research continues, particularly efforts to elucidate key aspects of structure, a clear path to a successful disease modifying therapy can be envisioned. Ensuring that lessons learned from recent, late-stage clinical failures are applied appropriately throughout therapeutic development will further enable the likelihood of a successful therapy in the near-term.
Collapse
Affiliation(s)
- Erika N Cline
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Maíra Assunção Bicca
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Kirsten L Viola
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - William L Klein
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
59
|
Wang X, Zhang J, Zhou L, Xu B, Ren X, He K, Nie L, Li X, Liu J, Yang X, Yuan J. Long-term iron exposure causes widespread molecular alterations associated with memory impairment in mice. Food Chem Toxicol 2019; 130:242-252. [PMID: 31136779 DOI: 10.1016/j.fct.2019.05.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/09/2019] [Accepted: 05/24/2019] [Indexed: 12/14/2022]
Abstract
Limited literature available indicates the neurotoxic effects of excessive iron, however, a deep understanding of iron neurotoxicity needs to be developed. In this study, we evaluated the toxic effects of excessive iron on learning and cognitive function in long-term iron exposure (oral, 10 mg/L, 6 months) of mice by behavioral tests including novel object recognition test, step-down passive avoidance test and Morris water maze test, and further analyzed differential expression of hippocampal proteins. The behavioral tests consistently showed that iron treatment caused cognitive defects of the mice. Proteomic analysis revealed 66 differentially expressed hippocampal proteins (30 increased and 36 decreased) in iron-treated mice as compared with the control ones. Bioinformatics analysis showed that the dysregulated proteins mainly included: synapse-associated proteins (i.e. synaptosomal-associated protein 25 (SNAP25), complexin-1 (CPLX1), vesicle-associated membrane protein 2 (VAMP2), neurochondrin (NCDN)); mitochondria-related proteins (i.e. ADP/ATP translocase 1 (SLC25A4), 14-3-3 protein zeta/delta (YWHAZ)); cytoskeleton proteins (i.e. neurofilament light polypeptide (NEFL), tubulin beta-2B chain (TUBB2B), tubulin alpha-4A chain (TUBA4A)). The findings suggest that the dysregulations of synaptic, mitochondrial, and cytoskeletal proteins may be involved in iron-triggered memory impairment. This study provides new insights into the molecular mechanisms of iron neurotoxicity.
Collapse
Affiliation(s)
- Xian Wang
- Department of Occupational and Environmental Health and Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China; Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, PR China
| | - Jiafei Zhang
- Department of Occupational and Environmental Health and Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China; Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, PR China
| | - Li Zhou
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, PR China
| | - Benhong Xu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, PR China
| | - Xiaohu Ren
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, PR China
| | - Kaiwu He
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, PR China
| | - Lulin Nie
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, PR China
| | - Xiao Li
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, PR China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, PR China.
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, PR China.
| | - Jing Yuan
- Department of Occupational and Environmental Health and Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China.
| |
Collapse
|
60
|
Shanbhag NM, Evans MD, Mao W, Nana AL, Seeley WW, Adame A, Rissman RA, Masliah E, Mucke L. Early neuronal accumulation of DNA double strand breaks in Alzheimer's disease. Acta Neuropathol Commun 2019; 7:77. [PMID: 31101070 PMCID: PMC6524256 DOI: 10.1186/s40478-019-0723-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 02/06/2023] Open
Abstract
The maintenance of genomic integrity is essential for normal cellular functions. However, it is difficult to maintain over a lifetime in postmitotic cells such as neurons, in which DNA damage increases with age and is exacerbated by multiple neurological disorders, including Alzheimer's disease (AD). Here we used immunohistochemical staining to detect DNA double strand breaks (DSBs), the most severe form of DNA damage, in postmortem brain tissues from patients with mild cognitive impairment (MCI) or AD and from cognitively unimpaired controls. Immunostaining for γH2AX-a post-translational histone modification that is widely used as a marker of DSBs-revealed increased proportions of γH2AX-labeled neurons and astrocytes in the hippocampus and frontal cortex of MCI and AD patients, as compared to age-matched controls. In contrast to the focal pattern associated with DSBs, some neurons and glia in humans and mice showed diffuse pan-nuclear patterns of γH2AX immunoreactivity. In mouse brains and primary neuronal cultures, such pan-nuclear γH2AX labeling could be elicited by increasing neuronal activity. To assess whether pan-nuclear γH2AX represents DSBs, we used a recently developed technology, DNA damage in situ ligation followed by proximity ligation assay, to detect close associations between γH2AX sites and free DSB ends. This assay revealed no evidence of DSBs in neurons or astrocytes with prominent pan-nuclear γH2AX labeling. These findings suggest that focal, but not pan-nuclear, increases in γH2AX immunoreactivity are associated with DSBs in brain tissue and that these distinct patterns of γH2AX formation may have different causes and consequences. We conclude that AD is associated with an accumulation of DSBs in vulnerable neuronal and glial cell populations from early stages onward. Because of the severe adverse effects this type of DNA damage can have on gene expression, chromatin stability and cellular functions, DSBs could be an important causal driver of neurodegeneration and cognitive decline in this disease.
Collapse
Affiliation(s)
- Niraj M Shanbhag
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Mark D Evans
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
| | - Wenjie Mao
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
| | - Alissa L Nana
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Anthony Adame
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, 92093, USA
- Present address: Division of Neuroscience, National Institute on Aging, Bethesda, MD, 20892, USA
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
61
|
Matias I, Morgado J, Gomes FCA. Astrocyte Heterogeneity: Impact to Brain Aging and Disease. Front Aging Neurosci 2019; 11:59. [PMID: 30941031 PMCID: PMC6433753 DOI: 10.3389/fnagi.2019.00059] [Citation(s) in RCA: 223] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/01/2019] [Indexed: 12/13/2022] Open
Abstract
Astrocytes, one of the largest glial cell population in the central nervous system (CNS), play a key function in several events of brain development and function, such as synapse formation and function, control of neurotransmitters release and uptake, production of trophic factors and control of neuronal survival. Initially described as a homogenous population, several evidences have pointed that astrocytes are highly heterogeneous, both morphologically and functionally, within the same region, and across different brain regions. Recent findings suggest that the heterogeneity in the expression profile of proteins involved in astrocyte function may predict the selective vulnerability of brain regions to specific diseases, as well as to the age-related cognitive decline. However, the molecular mechanisms underlying these changes, either in aging as well as in brain disease are scarce. Neuroinflammation, a hallmark of several neurodegenerative diseases and aging, is reported to have a dubious impact on glial activation, as these cells release pro- and anti-inflammatory cytokines and chemokines, anti-oxidants, free radicals, and neurotrophic factors. Despite the emerging evidences supporting that reactive astrocytes have a duality in their phenotype, neurotoxic or neuroprotective properties, depending on the age and stimuli, the underlying mechanisms of their activation, cellular interplays and the impact of regional astrocyte heterogeneity are still a matter of discussion. In this review article, we will summarize recent findings on astrocyte heterogeneity and phenotypes, as well as their likely impact for the brain function during aging and neural diseases. We will focus on the molecules and mechanisms triggered by astrocyte to control synapse formation in different brain regions. Finally, we will discuss new evidences on how the modulation of astrocyte phenotype and function could impact the synaptic deficits and glial dysfunction present in aging and pathological states.
Collapse
Affiliation(s)
- Isadora Matias
- Laboratory of Cellular Neurobiology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Morgado
- Laboratory of Cellular Neurobiology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flávia Carvalho Alcantara Gomes
- Laboratory of Cellular Neurobiology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
62
|
Mouchard A, Boutonnet MC, Mazzocco C, Biendon N, Macrez N. ApoE-fragment/Aβ heteromers in the brain of patients with Alzheimer's disease. Sci Rep 2019; 9:3989. [PMID: 30850702 PMCID: PMC6408522 DOI: 10.1038/s41598-019-40438-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/13/2019] [Indexed: 11/09/2022] Open
Abstract
Identification of endogenous pathological amyloid β peptides (Aβ) forms in the brains of patients with Alzheimer’s disease (AD) is still unclear. In healthy brain, Aβ can associate with Apolipoprotein E (ApoE) which is involved in its metabolism and clearance. In the brain of patients with AD, ApoE is cleaved and produces ApoE fragments. We studied the forms of Aβ and their interaction with the ApoE fragments in post-mortem brains from control and AD patients by western blots and co-immunoprecipitation. Three Aβ-containing peptides and three ApoE fragments were specifically found in the brain of AD patients. Co-immunoprecipitations showed that ApoE fragments and Aβ1–42 peptides are co-partners in heteromers of 18 and 16 kDa while ApoE-fragments and Aβ peptides of 12 kDa did not interact with each other. Formation of the 18 kDa ApoE-fragment/Aβ heteromers is specifically increased in ApoE4 carriers and is a strong brain marker of AD while 16 kDa ApoE-fragment/Aβ and Aβ 12 kDa correlate to memory deficit. These data show that in patients with AD, ApoE fragmentation generates peptides that trap Aβ in the brain. Inhibiting the fragmentation or targeting ApoE fragments could be exploited to define strategies to detect or reverse AD.
Collapse
Affiliation(s)
- Amandine Mouchard
- Bordeaux University, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France
| | - Marie-Charlotte Boutonnet
- Bordeaux University, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France
| | - Claire Mazzocco
- Bordeaux University, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France
| | - Nathalie Biendon
- Bordeaux University, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France
| | - Nathalie Macrez
- Bordeaux University, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France. .,CNRS, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France.
| | | |
Collapse
|
63
|
Zhao Y, Jaber VR, LeBeauf A, Sharfman NM, Lukiw WJ. microRNA-34a (miRNA-34a) Mediated Down-Regulation of the Post-synaptic Cytoskeletal Element SHANK3 in Sporadic Alzheimer's Disease (AD). Front Neurol 2019; 10:28. [PMID: 30792687 PMCID: PMC6374620 DOI: 10.3389/fneur.2019.00028] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022] Open
Abstract
Integrating a combination of bioinformatics, microRNA microfluidic arrays, ELISA analysis, LED Northern, and transfection-luciferase reporter assay data using human neuronal-glial (HNG) cells in primary culture we have discovered a set of up-regulated microRNAs (miRNAs) linked to a small family of down-regulated messenger RNAs (mRNAs) within the superior temporal lobe neocortex (Brodmann A22) of sporadic Alzheimer's disease (AD) brain. At the level of mRNA abundance, the expression of a significant number of human brain genes found to be down-regulated in sporadic AD neocortex appears to be due to the increased abundance of a several brain-abundant inducible miRNAs. These up-regulated miRNAs—including, prominently, miRNA-34a—have complimentary RNA sequences in the 3′ untranslated-region (3′-UTR) of their target-mRNAs that results in the pathological down-regulation in the expression of important brain genes. An up-regulated microRNA-34a, already implicated in age-related inflammatory-neurodegeneration–appears to down-regulate key mRNA targets involved in synaptogenesis and synaptic-structure, distinguishing neuronal deficits associated with AD neuropathology. One significantly down-regulated post-synaptic element in AD is the proline-rich SH3 and multiple-ankyrin-repeat domain SHANK3 protein. Bioinformatics, microRNA array analysis and SHANK3-mRNA-3′UTR luciferase-reporter assay confirmed the importance of miRNA-34a in the regulation of SHANK3 expression in HNG cells. This paper reports on recent studies of a miRNA-34a-up-regulation coupled to SHANK3 mRNA down-regulation in sporadic AD superior-temporal lobe compared to age-matched controls. These findings further support our hypothesis of an altered miRNA-mRNA coupled signaling network in AD, much of which is supported, and here reviewed, by recently reported experimental-findings in the scientific literature.
Collapse
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States.,Department of Anatomy and Cell Biology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Vivian R Jaber
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States
| | - Ayrian LeBeauf
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States
| | - Nathan M Sharfman
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States
| | - Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States.,Department of Ophthalmology, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States.,Department of Neurology, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States
| |
Collapse
|
64
|
W. Pilkington IV A, Legleiter J. Challenges in understanding the structure/activity relationship of Aβ oligomers. AIMS BIOPHYSICS 2019. [DOI: 10.3934/biophy.2019.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
65
|
Hassen GW, Kesner L, Stracher A, Shulman A, Rockenstein E, Mante M, Adame A, Overk C, Rissman RA, Masliah E. Effects of Novel Calpain Inhibitors in Transgenic Animal Model of Parkinson's disease/dementia with Lewy bodies. Sci Rep 2018; 8:18083. [PMID: 30591714 PMCID: PMC6308237 DOI: 10.1038/s41598-018-35729-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are neurodegenerative disorders of the aging population characterized by the accumulation of α-synuclein (α-syn). The mechanisms triggering α-syn toxicity are not completely understood, however, c-terminus truncation of α-syn by proteases such as calpain may have a role. Therefore, inhibition of calpain may be of value. The main objective of this study was to evaluate the effects of systemically administered novel low molecular weight calpain inhibitors on α-syn pathology in a transgenic mouse model. For this purpose, non-tg and α-syn tg mice received the calpain inhibitors - Gabadur, Neurodur or a vehicle, twice a day for 30 days. Immunocytochemical analysis showed a 60% reduction in α-syn deposition using Gabadur and a 40% reduction using Neurodur with a concomitant reduction in c-terminus α-syn and improvements in neurodegeneration. Western blot analysis showed a 77% decrease in α-spectrin breakdown products (SBDPs) SBDPs with Gabadur and 63% reduction using Neurodur. There was a 65% reduction in the active calpain form with Gabadur and a 45% reduction with Neurodur. Moreover, treatment with calpain inhibitors improved activity performance of the α-syn tg mice. Taken together, this study suggests that calpain inhibition might be considered in the treatment of synucleinopathies.
Collapse
Affiliation(s)
- Getaw Worku Hassen
- Department of Emergency Medicine, New York Medical College, Metropolitan Hospital Center, New York, USA
- Center for Drug Delivery Research, SUNY Downstate Medical Center, New York, USA
| | - Leo Kesner
- Department of Emergency Medicine, New York Medical College, Metropolitan Hospital Center, New York, USA
| | - Alfred Stracher
- Department of Emergency Medicine, New York Medical College, Metropolitan Hospital Center, New York, USA
| | - Abraham Shulman
- Department of Otorhinolaryngology, SUNY Downstate Medical Center, New York, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, California, 92093-0624, USA
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, La Jolla, California, 92093-0624, USA
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, California, 92093-0624, USA
| | - Cassia Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, California, 92093-0624, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, California, 92093-0624, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, California, 92093-0624, USA.
- Division of Neurosciences and Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
66
|
Kakinen A, Sun Y, Javed I, Faridi A, Pilkington EH, Faridi P, Purcell AW, Zhou R, Ding F, Lin S, Chun Ke P, Davis TP. Physical and Toxicological Profiles of Human IAPP Amyloids and Plaques. Sci Bull (Beijing) 2018; 64:26-35. [PMID: 30662791 DOI: 10.1016/j.scib.2018.11.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although much has been learned about the fibrillization kinetics, structure and toxicity of amyloid proteins, the properties of amyloid fibrils beyond the saturation phase are often perceived as chemically and biologically inert, despite evidence suggesting otherwise. To fill this knowledge gap, we examined the physical and biological characteristics of human islet amyloid polypeptide (IAPP) fibrils that were aged up to two months. Not only did aging decrease the toxicity of IAPP fibrils, but the fibrils also sequestered fresh IAPP and suppressed their toxicity in an embryonic zebrafish model. The mechanical properties of IAPP fibrils in different aging stages were probed by atomic force microscopy and sonication, which displayed comparable stiffness but age-dependent fragmentation, followed by self-assembly of such fragments into the largest lamellar amyloid structures reported to date. The dynamic structural and toxicity profiles of amyloid fibrils and plaques suggest that they play active, long-term roles in cell degeneration and may be a therapeutic target for amyloid diseases.
Collapse
Affiliation(s)
- Aleksandr Kakinen
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| | - Yunxiang Sun
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Ibrahim Javed
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia.,College of Environmental Science and Engineering, Shanghai Institute of Pollution Control and Ecological Security, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Ava Faridi
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| | - Emily H Pilkington
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| | - Pouya Faridi
- Department of Biochemistry and Molecular Biology, Infection and Immunity Program & Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology, Infection and Immunity Program & Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Ruhong Zhou
- IBM Thomas J. Watson Research Center, Yorktown Heights, New York, 10598, USA.,Department of Chemistry, Columbia University, New York, New York, 10027, USA
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Sijie Lin
- College of Environmental Science and Engineering, Shanghai Institute of Pollution Control and Ecological Security, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| |
Collapse
|
67
|
Chai YL, Chong JR, Weng J, Howlett D, Halsey A, Lee JH, Attems J, Aarsland D, Francis PT, Chen CP, Lai MKP. Lysosomal cathepsin D is upregulated in Alzheimer's disease neocortex and may be a marker for neurofibrillary degeneration. Brain Pathol 2018; 29:63-74. [PMID: 30051532 DOI: 10.1111/bpa.12631] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/12/2018] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by accumulation of β-amyloid plaques (AP) and neurofibrillary tangles (NFT) in the cortex, together with synaptic loss and amyloid angiopathy. Perturbations in the brain lysosomal system, including the cathepsin family of proteases, have been implicated in AD where they may be involved in proteolytic clearance of misfolded and abnormally aggregated peptides. However, the status of cathepsin D (catD) is unclear in Lewy body dementia, the second most common form of neurodegenerative dementia after AD, and characterized by Lewy bodies (LB) containing aggregated α-synuclein. Furthermore, earlier reports of catD changes in AD have not been entirely consistent. We measured CatD immunoreactivities in the temporal (Brodmann area BA21) and parietal (BA40) cortices of well characterized AD brains as well as two clinical subtypes of Lewy body dementia, namely Parkinson disease dementia (PDD) and dementia with Lewy bodies (DLB), known to show varying degrees of concomitant AD pathology. Increased catD immunoreactivities in AD were found for both neocortical regions measured, where they also correlated with neuropathological NFT scores and phosphorylated pSer396 tau burden, and appeared to co-localize at least partly to NFT-containing neurons. In contrast, catD was increased only in BA40 in DLB and not at all in PDD, did not correlate with LB scores, and did not appreciably co-localize with α-synuclein inclusions. Our study suggests that catD upregulation may be an adaptive response to AD-related processes leading to neurofibrillary degeneration, but may not be directly associated with formation of α-synuclein inclusions in Lewy body dementia.
Collapse
Affiliation(s)
- Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory Aging and Cognition Centre, National University Health System, Kent Ridge, Singapore
| | - Jiaju Weng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - David Howlett
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Andrea Halsey
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Jasinda H Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Johannes Attems
- Institute of Neuroscience, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Dag Aarsland
- Department NVS, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institute, Huddinge, Sweden
| | - Paul T Francis
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory Aging and Cognition Centre, National University Health System, Kent Ridge, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.,Memory Aging and Cognition Centre, National University Health System, Kent Ridge, Singapore.,Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| |
Collapse
|
68
|
Zhang H, Therriault J, Kang MS, Ng KP, Pascoal TA, Rosa-Neto P, Gauthier S. Cerebrospinal fluid synaptosomal-associated protein 25 is a key player in synaptic degeneration in mild cognitive impairment and Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2018; 10:80. [PMID: 30115118 PMCID: PMC6097333 DOI: 10.1186/s13195-018-0407-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/12/2018] [Indexed: 12/02/2022]
Abstract
Background There is accumulating evidence that synaptic loss precedes neuronal loss and correlates best with impaired memory formation in Alzheimer’s disease (AD). Cerebrospinal fluid (CSF) synaptosomal-associated protein 25 (SNAP-25) is a newly discovered marker indicating synaptic damage. We here test CSF SNAP-25 and SNAP-25/amyloid-β42 (Aβ42) ratio as a diagnostic marker for predicting cognitive decline and brain structural change in the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database. Methods We stratified 139 participants from the ADNI database into cognitively normal (CN; n = 52), stable mild cognitive impairment (sMCI; n = 22), progressive MCI (pMCI; n = 47), and dementia due to AD (n = 18). Spearman correlation was performed to test the relationships between biomarkers. Overall diagnostic accuracy (area under the curve (AUC)) was obtained from receiver operating curve (ROC) analyses. Cox proportional hazard models tested the effect of CSF SNAP-25 and SNAP-25/Aβ42 measures on the conversion from MCI to AD. Relationships between the CSF SNAP-25 levels, SNAP-25/Aβ42 ratio, and diagnostic groups were tested with linear regressions. Linear mixed-effects models and linear regression models were used to evaluate CSF SNAP-25 and SNAP-25/Aβ42 as predictors of AD features, including cognition measured by the Mini-Mental State Examination (MMSE) and brain structure and white matter hyperintensity (WMH) measured by magnetic resonance imaging (MRI). Results CSF SNAP-25 and SNAP-25/Aβ42 were increased in patients with pMCI and AD compared with CN, and in pMCI and AD compared with sMCI. Cognitively normal subjects who progressed to MCI or AD during follow-up had increased SNAP-25/Aβ42 ratio compared with nonprogressors. CSF SNAP-25, especially SNAP-25/Aβ42, offers diagnostic utility for pMCI and AD. CSF SNAP-25 and SNAP-25/Aβ42 significantly predicted conversion from MCI to AD. In addition, elevated SNAP-25/Aβ42 ratio was associated with the rate of hippocampal atrophy in pMCI and the rate of change of cognitive impairment in CN over the follow-up period. Conclusions These data suggest that both CSF SNAP-25 and SNAP-25/Aβ42 ratio are already increased at the early clinical stage of AD, and indicate the promise of CSF SNAP-25 and SNAP-25/Aβ42 ratio as diagnostic and prognostic biomarkers for the earliest symptomatic stage of AD.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Joseph Therriault
- The McGill University Research Centre for Studies in Aging, McGill University, Montreal, Canada
| | - Min Su Kang
- The McGill University Research Centre for Studies in Aging, McGill University, Montreal, Canada
| | - Kok Pin Ng
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Tharick A Pascoal
- The McGill University Research Centre for Studies in Aging, McGill University, Montreal, Canada
| | - Pedro Rosa-Neto
- The McGill University Research Centre for Studies in Aging, McGill University, Montreal, Canada
| | - Serge Gauthier
- The McGill University Research Centre for Studies in Aging, McGill University, Montreal, Canada.
| | | |
Collapse
|
69
|
Alexander NJ, Rau AR, Jimenez VA, Daunais JB, Grant KA, McCool BA. SNARE Complex-Associated Proteins in the Lateral Amygdala of Macaca mulatta Following Long-Term Ethanol Drinking. Alcohol Clin Exp Res 2018; 42:1661-1673. [PMID: 29944190 DOI: 10.1111/acer.13821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/21/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent work with long-term ethanol (EtOH) self-administration in nonhuman primate models has revealed a complex array of behavioral and physiological effects that closely mimic human alcohol abuse. Detailed neurophysiological analysis in these models suggests a myriad of pre- and postsynaptic neurobiological effects that may contribute to the behavioral manifestations of long-term EtOH drinking. The molecular mechanisms regulating presynaptic effects of this chronic EtOH exposure are largely unknown. To this end, we analyzed the effects of long-term EtOH self-administration on the levels of presynaptic SNARE complex proteins in Macaca mulatta basolateral amygdala, a brain region known to regulate both aversive and reward-seeking behaviors. METHODS Basolateral amygdala samples from control and EtOH-drinking male and female monkeys were processed. Total basolateral amygdala protein was analyzed by Western blotting using antibodies directed against both core SNARE and SNARE-associated proteins. We also performed correlational analyses between protein expression levels and a number of EtOH drinking parameters, including lifetime grams of EtOH consumed, preference, and blood alcohol concentration. RESULTS Significant interactions or main effects of sex/drinking were seen for a number of SNARE core and SNARE-associated proteins. Across the range of EtOH-drinking phenotypes, SNAP25 and Munc13-1 proteins levels were significantly different between males and females, and Munc13-2 levels were significantly lower in animals with a history of EtOH drinking. A separate analysis of very heavy-drinking individuals revealed significant decreases in Rab3c (females) and complexin 2 (males). CONCLUSIONS Protein expression analysis of basolateral amygdala total protein from controls and animals following long-term EtOH self-administration suggests a number of alterations in core SNARE or SNARE-associated components that could dramatically alter presynaptic function. A number of proteins or multiprotein components were also correlated with EtOH drinking behavior, which suggest a potentially heritable role for presynaptic SNARE proteins.
Collapse
Affiliation(s)
- Nancy J Alexander
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Andrew R Rau
- Department of Behavioral Neuroscience, Oregon National Primate Research Center, Oregon Health Sciences University, Portland, Oregon
| | - Vanessa A Jimenez
- Department of Behavioral Neuroscience, Oregon National Primate Research Center, Oregon Health Sciences University, Portland, Oregon
| | - James B Daunais
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Kathleen A Grant
- Department of Behavioral Neuroscience, Oregon National Primate Research Center, Oregon Health Sciences University, Portland, Oregon
| | - Brian A McCool
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
70
|
Lee CT, Terentjev EM. Mechanisms and rates of nucleation of amyloid fibrils. J Chem Phys 2018; 147:105103. [PMID: 28915747 DOI: 10.1063/1.4995255] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The classical nucleation theory finds the rate of nucleation proportional to the monomer concentration raised to the power, which is the "critical nucleus size," nc. The implicit assumption, that amyloids nucleate in the same way, has been recently challenged by an alternative two-step mechanism, when the soluble monomers first form a metastable aggregate (micelle) and then undergo conversion into the conformation rich in β-strands that are able to form a stable growing nucleus for the protofilament. Here we put together the elements of extensive knowledge about aggregation and nucleation kinetics, using a specific case of Aβ1-42 amyloidogenic peptide for illustration, to find theoretical expressions for the effective rate of amyloid nucleation. We find that at low monomer concentrations in solution and also at low interaction energy between two peptide conformations in the micelle, the nucleation occurs via the classical route. At higher monomer concentrations, and a range of other interaction parameters between peptides, the two-step "aggregation-conversion" mechanism of nucleation takes over. In this regime, the effective rate of the process can be interpreted as a power of monomer concentration in a certain range of parameters; however, the exponent is determined by a complicated interplay of interaction parameters and is not related to the minimum size of the growing nucleus (which we find to be ∼7-8 for Aβ1-42).
Collapse
Affiliation(s)
- Cheng-Tai Lee
- Cavendish Laboratory, University of Cambridge, J.J. Thomson Avenue, Cambridge CB3 0HE, United Kingdom
| | - Eugene M Terentjev
- Cavendish Laboratory, University of Cambridge, J.J. Thomson Avenue, Cambridge CB3 0HE, United Kingdom
| |
Collapse
|
71
|
Yu L, Liu Y, Yang H, Zhu X, Cao X, Gao J, Zhao H, Xu Y. PSD-93 Attenuates Amyloid-β-Mediated Cognitive Dysfunction by Promoting the Catabolism of Amyloid-β. J Alzheimers Dis 2018; 59:913-927. [PMID: 28697571 DOI: 10.3233/jad-170320] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Amyloid-β (Aβ) is a key neuropathological hallmark of Alzheimer's disease (AD). Postsynaptic density protein 93 (PSD-93) is a key scaffolding protein enriched at postsynaptic sites. The aim of the present study was to examine whether PSD-93 overexpression could alleviate Aβ-induced cognitive dysfunction in APPswe/PS1dE9 (APP/PS1) mice by reducing Aβ levels in the brain. The level of PSD-93 was significantly decreased in the hippocampus of 6-month-old APP/PS1 mice compared with that in wild-type mice. Following lentivirus-mediated PSD-93 overexpression, cognitive function, synaptic function, and amyloid burden were investigated. The open field test, Morris water maze test, and fear condition test revealed that PSD-93 overexpression ameliorated spatial memory deficits in APP/PS1 mice. The facilitation of long-term potentiation induction was observed in APP/PS1 mice after PSD-93 overexpression. The expression of somatostatin receptor 4 (SSTR4) and neprilysin was increased, while the amyloid plaque load and Aβ levels were decreased in the brains of APP/PS1 mice. Moreover, PSD-93 interacted with SSTR4 and affected the level of SSTR4 on cell membrane, which was associated with the ubiquitination. Together, these findings suggest that PSD-93 attenuates spatial memory deficits and decreases amyloid levels in APP/PS1 mice, which might be associated with Aβ catabolism, and overexpression of PSD-93 might be a potential therapy for AD.
Collapse
Affiliation(s)
- Linjie Yu
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, P. R. China.,The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China
| | - Yi Liu
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, P. R. China.,The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China
| | - Hui Yang
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, P. R. China.,Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Xiaolei Zhu
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, P. R. China.,The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, P. R. China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, P. R. China.,The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, P. R. China
| | - Jun Gao
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing, P. R. China
| | - Hui Zhao
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, P. R. China.,The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, P. R. China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, P. R. China.,The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, P. R. China.,Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, P. R. China
| |
Collapse
|
72
|
nArgBP2-SAPAP-SHANK, the core postsynaptic triad associated with psychiatric disorders. Exp Mol Med 2018; 50:1-9. [PMID: 29628500 PMCID: PMC5938024 DOI: 10.1038/s12276-017-0018-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 11/23/2022] Open
Abstract
Despite the complex genetic architecture, a broad spectrum of psychiatric disorders can still be caused by mutation(s) in the same gene. These disorders are interrelated with overlapping causative mechanisms including variations in the interaction among the risk-associated proteins that may give rise to the specific spectrum of each disorder. Additionally, multiple lines of evidence implicate an imbalance between excitatory and inhibitory neuronal activity (E/I imbalance) as the shared key etiology. Thus, understanding the molecular mechanisms underlying E/I imbalance provides essential insight into the etiology of these disorders. One important class of candidate risk genes is the postsynaptic scaffolding proteins, such as nArgBP2, SAPAP, and SHANK that regulate the actin cytoskeleton in dendritic spines of excitatory synapses. This review will cover and discuss recent studies that examined how these proteins, especially nArgBP2, are associated with psychiatric disorders. Next, we propose a possibility that variations in the interaction among these proteins in a specific brain region might contribute to the onset of diverse phenotypes of psychiatric disorders. The assembly of scaffolding proteins, key regulators of many signaling pathways, found in the brain’s synapses underpin a diverse range of neuropsychiatric disorders. Sunghoe Chang and colleagues from Seoul National University, South Korea, review how these postsynaptic proteins regulate the cellular cytoskeleton in nerve cell protrusions to maintain the balance between excitatory and inhibitory inputs in the brain. They discuss how perturbations in three particular proteins can cause an imbalance in synaptic signals that leads to conditions such as bipolar disorder, schizophrenia and autism. The authors propose that these proteins form a “core scaffolding triad” and interact in different ways to cause different mental illnesses. Dysregulation of these proteins could explain how mutations in the same genes, depending on whether they boost or decrease gene expression, contribute to the onset of diverse psychiatric disorders.
Collapse
|
73
|
Chen F, He Y, Wang P, Wei P, Feng H, Rao Y, Shi J, Tian J. Curcumin can influence synaptic dysfunction in APPswe/PS1dE9 mice. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2018. [DOI: 10.1016/j.jtcms.2018.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
74
|
Cheignon C, Tomas M, Bonnefont-Rousselot D, Faller P, Hureau C, Collin F. Oxidative stress and the amyloid beta peptide in Alzheimer's disease. Redox Biol 2018; 14:450-464. [PMID: 29080524 PMCID: PMC5680523 DOI: 10.1016/j.redox.2017.10.014] [Citation(s) in RCA: 1336] [Impact Index Per Article: 222.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/14/2017] [Accepted: 10/17/2017] [Indexed: 01/12/2023] Open
Abstract
Oxidative stress is known to play an important role in the pathogenesis of a number of diseases. In particular, it is linked to the etiology of Alzheimer's disease (AD), an age-related neurodegenerative disease and the most common cause of dementia in the elderly. Histopathological hallmarks of AD are intracellular neurofibrillary tangles and extracellular formation of senile plaques composed of the amyloid-beta peptide (Aβ) in aggregated form along with metal-ions such as copper, iron or zinc. Redox active metal ions, as for example copper, can catalyze the production of Reactive Oxygen Species (ROS) when bound to the amyloid-β (Aβ). The ROS thus produced, in particular the hydroxyl radical which is the most reactive one, may contribute to oxidative damage on both the Aβ peptide itself and on surrounding molecule (proteins, lipids, …). This review highlights the existing link between oxidative stress and AD, and the consequences towards the Aβ peptide and surrounding molecules in terms of oxidative damage. In addition, the implication of metal ions in AD, their interaction with the Aβ peptide and redox properties leading to ROS production are discussed, along with both in vitro and in vivo oxidation of the Aβ peptide, at the molecular level.
Collapse
Affiliation(s)
- C Cheignon
- LCC (Laboratoire de Chimie de Coordination), CNRS UPR 8241, 205 route de Narbonne, 31062 Toulouse Cedex 09, France; Université de Toulouse; UPS, INPT, 31077 Toulouse, France
| | - M Tomas
- LCC (Laboratoire de Chimie de Coordination), CNRS UPR 8241, 205 route de Narbonne, 31062 Toulouse Cedex 09, France; Université de Toulouse; UPS, INPT, 31077 Toulouse, France
| | - D Bonnefont-Rousselot
- Department of Metabolic Biochemistry, La Pitié Salpêtrière-Charles Foix University Hospital (AP-HP), Paris, France; Department of Biochemistry, Faculty of Pharmacy, Paris Descartes University, Paris, France; CNRS UMR8258 - INSERM U1022, Faculty of Pharmacy, Paris Descartes University, Paris, France
| | - P Faller
- Biometals and Biology Chemistry, Institut de Chimie (CNRS UMR 7177), University of Strasbourg, 4 rue B. Pascal, 67081 Strasbourg Cedex, France
| | - C Hureau
- LCC (Laboratoire de Chimie de Coordination), CNRS UPR 8241, 205 route de Narbonne, 31062 Toulouse Cedex 09, France; Université de Toulouse; UPS, INPT, 31077 Toulouse, France
| | - F Collin
- LCC (Laboratoire de Chimie de Coordination), CNRS UPR 8241, 205 route de Narbonne, 31062 Toulouse Cedex 09, France; Université de Toulouse; UPS, INPT, 31077 Toulouse, France.
| |
Collapse
|
75
|
Zhang S, Chai R, Yang YY, Guo SQ, Wang S, Guo T, Xu SF, Zhang YH, Wang ZY, Guo C. Chronic diabetic states worsen Alzheimer neuropathology and cognitive deficits accompanying disruption of calcium signaling in leptin-deficient APP/PS1 mice. Oncotarget 2018; 8:43617-43634. [PMID: 28467789 PMCID: PMC5546429 DOI: 10.18632/oncotarget.17116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/03/2017] [Indexed: 02/06/2023] Open
Abstract
The coincidences between Alzheimer’s disease (AD) and type 2 diabetes mellitus (T2DM) are so compelling that it is attractive to speculate that diabetic conditions might aggravate AD pathologies by calcium dysfunction, although the understanding of the molecular mechanisms involved remains elusive. The present work was undertaken to investigate whether calcium dyshomeostasis is associated with the exacerbated Alzheimer-like cognitive dysfunction observed in diabetic conditions in APP/PS1-ob/ob mice, which were generated by crossing ob/ob mice with APP/PS1 mice. We confirmed that the diabetic condition can aggravate not only Aβ deposition but also tau phosphorylation, synaptic loss, neuronal death, and inflammation, exacerbating cognitive impairment in AD mice. More importantly, we found that the diabetic condition dramatically elevated calcium levels in APP/PS1 mice, thereby stimulating the phosphorylation of the calcium-dependent kinases. Our findings suggest that controlling over-elevation of intracellular calcium may provide novel insights for approaching AD in diabetic patients and delaying AD progression.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Rui Chai
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Ying-Ying Yang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Shi-Qi Guo
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Shan Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Tian Guo
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Shuang-Feng Xu
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yan-Hui Zhang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
76
|
Zhao Y, Lukiw WJ. Microbiome-Mediated Upregulation of MicroRNA-146a in Sporadic Alzheimer's Disease. Front Neurol 2018; 9:145. [PMID: 29615954 PMCID: PMC5867462 DOI: 10.3389/fneur.2018.00145] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 02/27/2018] [Indexed: 12/13/2022] Open
Abstract
The first indication of a potential mechanistic link between the pathobiology of the human gastrointestinal (GI)-tract microbiome and its contribution to the pathogenetic mechanisms of sporadic Alzheimer's disease (AD) came a scant 4 years ago (1). Ongoing research continues to strengthen the hypothesis that neurotoxic microbial-derived components of the GI tract microbiome can cross aging GI tract and blood-brain barriers and contribute to progressive proinflammatory neurodegeneration, as exemplified by the AD-process. Of central interest in these recent investigations are the pathological roles played by human GI tract resident Gram-negative anaerobic bacteria and neurotropic viruses-two prominent divisions of GI tract microbiome-derived microbiota-which harbor considerable pathogenic potential. It is noteworthy that the first two well-studied microbiota-the GI tract abundant Gram-negative bacteria Bacteroides fragilis and the neurotropic herpes simplex virus-1 both share a final common pathway of NF-κB (p50/p65) activation and microRNA-146a induction with ensuing pathogenic stimulation of innate-immune and neuroinflammatory pathways. These appear to strongly contribute to the inflammation-mediated amyloidogenic neuropathology of AD. This communication: (i) will review recent research contributions that have expanded our understanding of the nature of the translocation of microbiome-derived neurotoxins-across biophysiological barriers; (ii) will assess multiple-recent investigations of the induction of the proinflammatory pathogenic microRNA-146a by these two prominent classes of human microbiota; and (iii) will discuss the role of molecular neurobiology and mechanistic contribution of polymicrobial infections to AD-type neuropathological change.
Collapse
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States
- Department of Anatomy and Cell Biology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States
- Department of Ophthalmology, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States
- Department of Neurology, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA, United States
| |
Collapse
|
77
|
Chong JR, Chai YL, Lee JH, Howlett D, Attems J, Ballard CG, Aarsland D, Francis PT, Chen CP, Lai MKP. Increased Transforming Growth Factor β2 in the Neocortex of Alzheimer's Disease and Dementia with Lewy Bodies is Correlated with Disease Severity and Soluble Aβ42 Load. J Alzheimers Dis 2018; 56:157-166. [PMID: 27911312 DOI: 10.3233/jad-160781] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Of the three transforming growth factor (TGF)-β isoforms known, TGFβ1 deficits have been widely reported in Alzheimer's disease (AD) and studied as a potential therapeutic target. In contrast, the status of TGFβ2, which has been shown to mediate amyloid-β (Aβ)-mediated neuronal death, are unclear both in AD and in Lewy body dementias (LBD) with differential neuritic plaque and neurofibrillary tangle burden. OBJECTIVE To measure neocortical TGFβ2 levels and their correlations with neuropathological and clinical markers of disease severity in a well-characterized cohort of AD as well as two clinical subtypes of LBD, dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD), known to manifest relatively high and low Aβ plaque burden, respectively. METHODS Postmortem samples from temporal cortex (BA21) were measured for TGFβ2 using a Luminex-based platform, and correlated with scores for neuritic plaques, neurofibrillary tangles, α-synuclein pathology, dementia severity (as measured by annual decline of Mini-Mental State Examination scores) as well as soluble and total fractions of brain Aβ42. RESULTS TGFβ2 was significantly increased in AD and DLB, but not in PDD. TGFβ2 also correlated with scores for neurofibrillary tangles, Lewy bodies (within the LBD group), dementia severity, and soluble Aβ42 concentration, but not with neuritic plaque scores, total Aβ42, or monomeric α-synuclein immunoreactivity. CONCLUSIONS TGFβ2 is increased in the temporal cortex of AD and DLB, and its correlations with neuropathological and clinical markers of disease severity as well as with soluble Aβ42 load suggest a potential pathogenic role in mediating the neurotoxicity of non-fibrillar Aβ. Our study also indicates the potential utility of targeting TGFβ2 in pharmacotherapeutic approaches to AD and DLB.
Collapse
Affiliation(s)
- Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jasinda H Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Howlett
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Johannes Attems
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Clive G Ballard
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Dag Aarsland
- Department of Neurobiology, Ward Sciences and Society, Karolinska Institute, Stockholm, Sweden.,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Paul T Francis
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| |
Collapse
|
78
|
Oliveira T, Starkweather A, Ramesh D, Fetta J, Kelly DL, Lyon DE, Sargent L. Putative mechanisms of cognitive decline with implications for clinical research and practice. Nurs Forum 2018; 53:10.1111/nuf.12247. [PMID: 29345733 PMCID: PMC6438756 DOI: 10.1111/nuf.12247] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multiple intrinsic and extrinsic mechanisms contribute to vulnerability of cognitive decline and nurses play a significant role in assisting individuals and families to use strategies for healthy cognitive aging. The objective of this narrative review is to provide a synthesis of the intrinsic and extrinsic mechanisms of cognitive decline and conditions that are associated with cognitive decline. Well-established intrinsic mechanisms of cognitive decline include aging, apolipoprotein E (APOE) ε4 carrier status, SORL1 mutations, neuroinflammation, mitochondrial dysfunction, amyloid deposition, and demyelination. Extrinsic risk factors include obesity, diabetes, hypertension, elevated lipid panel, metabolic syndrome, depression, traumatic brain injury, substance use, heart failure, and stroke. The various definitions of cognitive decline as well as the intrinsic and extrinsic factors that impact cognition as humans age should be incorporated in future clinical research studies. Nurses may use this information to help patients make lifestyle choices regarding cognitive health.
Collapse
Affiliation(s)
- Tadeu Oliveira
- School of Nursing, University of Connecticut, Storrs, CT
| | | | - Divya Ramesh
- School of Nursing, University of Connecticut, Storrs, CT
| | - Joseph Fetta
- School of Nursing, University of Connecticut, Storrs, CT
| | | | - Debra E. Lyon
- College of Nursing, University of Florida, Gainesville, FL
| | - Lana Sargent
- School of Nursing, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
79
|
A preclinical screen to evaluate pharmacotherapies for the treatment of agitation in dementia. Behav Pharmacol 2018; 28:199-206. [PMID: 28234659 DOI: 10.1097/fbp.0000000000000298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Agitation associated with dementia is frequently reported clinically but has received little attention in preclinical models of dementia. The current study used a 7PA2 CM intracerebroventricular injection model of Alzheimer's disease (AD) to assess acute memory impairment, and a bilateral intrahippocampal (IH) injection model of AD (aggregated Aβ1-42 injections) and a bilateral IH injection model of dementia with Lewy bodies (aggregated NAC61-95 injections) to assess chronic memory impairment in the rat. An alternating-lever cyclic-ratio schedule of operant responding was used for data collection, where incorrect lever perseverations measured executive function (memory) and running response rates (RRR) measured behavioral output (agitation). The results indicate that bilateral IH injections of Aβ1-42 and bilateral IH injections of NAC61-95 decreased memory function and increased RRRs, whereas intracerebroventricular injections of 7PA2 CM decreased memory function but did not increase RRRs. These findings show that using the aggregated peptide IH injection models of dementia to induce chronic neurotoxicity, memory decline was accompanied by elevated behavioral output. This demonstrates that IH peptide injection models of dementia provide a preclinical screen for pharmacological interventions used in the treatment of increased behavioral output (agitation), which also establish detrimental side effects on memory.
Collapse
|
80
|
Chung H, Crooks EJ, Ziliox M, Smith SO. Disaggregation of Aβ42 for Structural and Biochemical Studies. Methods Mol Biol 2018; 1777:321-330. [PMID: 29744845 DOI: 10.1007/978-1-4939-7811-3_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The amyloid-β (Aβ) peptides that form the amyloid fibrils associated with Alzheimer's disease are generated by sequential proteolysis of the amyloid precursor protein by β- and γ-secretase. The two predominant Aβ peptides, Aβ40 and Aβ42, differ by two amino acids, are soluble as monomers at low concentration (and/or low temperature) and are normally cleared from the brain parenchyma. In order to study the structure and assembly of these peptides, they are often synthesized using solid-phase peptide synthesis and purified. Here, we outline the method we use to prepare monomeric Aβ for structural and biochemical studies.
Collapse
Affiliation(s)
- Hyewon Chung
- Department of Ophthalmology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, South Korea
| | - Elliot J Crooks
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Martine Ziliox
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Steven O Smith
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
81
|
Ahmed R, Melacini G. A solution NMR toolset to probe the molecular mechanisms of amyloid inhibitors. Chem Commun (Camb) 2018; 54:4644-4652. [DOI: 10.1039/c8cc01380b] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A chemical exchange-based solution NMR toolset to probe the molecular mechanisms of amyloid inhibitors.
Collapse
Affiliation(s)
- Rashik Ahmed
- Department of Biochemistry and Biomedical Sciences
- McMaster University
- Hamilton
- Canada
| | - Giuseppe Melacini
- Department of Biochemistry and Biomedical Sciences
- McMaster University
- Hamilton
- Canada
- Department of Chemistry and Chemical Biology
| |
Collapse
|
82
|
Musardo S, Marcello E. Synaptic dysfunction in Alzheimer's disease: From the role of amyloid β-peptide to the α-secretase ADAM10. Eur J Pharmacol 2017. [DOI: 10.1016/j.ejphar.2017.06.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
83
|
Rego Â, Viana SD, Ribeiro CAF, Rodrigues-Santos P, Pereira FC. Monophosphoryl Lipid-A: A Promising Tool for Alzheimer's Disease Toll. J Alzheimers Dis 2017; 52:1189-202. [PMID: 27079716 DOI: 10.3233/jad-151183] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuroinflammation is a two-edged sword in Alzheimer's disease (AD). A certain degree of neuroinflammation is instrumental in the clearance of amyloid-β (Aβ) peptides by activated microglia, although a sustained neuroinflammation might accelerate Aβ deposition, thus fostering the neurodegenerative process and functional decline in AD. There is an increasing body of evidence suggesting that the innate immune system via Toll-like receptor 4 (TLR4) finely orchestrates the highly regulated inflammatory cascade that takes place in AD pathology. Herein we critically review pre-clinical (in vitro and in vivo approaches) and clinical studies showing that monophosphoryl lipid A (MPL), a partial TLR4 agonist, may have beneficial effect on AD physiopathology. The in vivo data elegantly showed that MPL enhanced Aβ plaque phagocytosis thus decreasing the number and the size of Aβ deposits and soluble Aβ in brain from APPswe/PS1 mice. Furthermore, MPL also improved their cognition. The mechanism underlying this MPL effect was proposed to be microglial activation by recruiting TLR4. Additionally, it was demonstrated that MPL increased the Aβ antibody titer and showed a safe profile in mice and primates, when used as a vaccine adjuvant. Clinical studies using MPL as an adjuvant in Aβ immunotherapy are currently ongoing. Overall, we argue that the TLR4 partial agonist MPL is a potentially safe and effective new pharmacological tool in AD.
Collapse
Affiliation(s)
- Ângela Rego
- Laboratório de Farmacologia e Terapêutica Experimental/IBILI, Faculdade de Medicina da Universidade de Coimbra, Portugal.,CNC.IBILI - Universidade de Coimbra, Coimbra, Portugal.,Centro Hospitalar do Porto, Largo Prof. Abel Salazar, Porto, Portugal
| | - Sofia D Viana
- Laboratório de Farmacologia e Terapêutica Experimental/IBILI, Faculdade de Medicina da Universidade de Coimbra, Portugal.,CNC.IBILI - Universidade de Coimbra, Coimbra, Portugal.,Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Farmácia, Portugal
| | - Carlos A Fontes Ribeiro
- Laboratório de Farmacologia e Terapêutica Experimental/IBILI, Faculdade de Medicina da Universidade de Coimbra, Portugal.,CNC.IBILI - Universidade de Coimbra, Coimbra, Portugal
| | - Paulo Rodrigues-Santos
- Instituto de Imunologia, Faculdade de Medicina da Universidade de Coimbra, Portugal.,Laboratório de Imunologia e Oncologia, Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Portugal
| | - Frederico C Pereira
- Laboratório de Farmacologia e Terapêutica Experimental/IBILI, Faculdade de Medicina da Universidade de Coimbra, Portugal.,CNC.IBILI - Universidade de Coimbra, Coimbra, Portugal
| |
Collapse
|
84
|
Hui S, Yang Y, Peng WJ, Sheng CX, Gong W, Chen S, Xu PP, Wang Z. Protective effects of Bushen Tiansui decoction on hippocampal synapses in a rat model of Alzheimer's disease. Neural Regen Res 2017; 12:1680-1686. [PMID: 29171433 PMCID: PMC5696849 DOI: 10.4103/1673-5374.217347] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2017] [Indexed: 12/26/2022] Open
Abstract
Bushen Tiansui decoction is composed of six traditional Chinese medicines: Herba Epimedii, Radix Polygoni multiflori, Plastrum testudinis, Fossilia Ossis Mastodi, Radix Polygalae, and Rhizoma Acorus tatarinowii. Because Bushen Tiansui decoction is effective against amyloid beta (Aβ) toxicity, we hypothesized that it would reduce hippocampal synaptic damage and improve cognitive function in Alzheimer's disease. To test this hypothesis, we used a previously established animal model of Alzheimer's disease, that is, microinjection of aggregated Aβ25-35 into the bilateral brain ventricles of Sprague-Dawley rats. We found that long-term (28 days) oral administration of Bushen Tiansui decoction (0.563, 1.688, and 3.375 g/mL; 4 mL/day) prevented synaptic loss in the hippocampus and increased the expression levels of synaptic proteins, including postsynaptic density protein 95, the N-methyl-D-aspartate receptor 2B subunit, and Shank1. These results suggested that Bushen Tiansui decoction can protect synapses by maintaining the expression of these synaptic proteins. Bushen Tiansui decoction also ameliorated measures reflecting spatial learning and memory deficits that were observed in the Morris water maze (i.e., increased the number of platform crossings and the amount of time spent in the target quadrant and decreased escape latency) following intraventricular injections of aggregated Aβ25-35 compared with those measures in untreated Aβ25-35-injected rats. Overall, these results provided evidence that further studies on the prevention and treatment of dementia with this traditional Chinese medicine are warranted.
Collapse
Affiliation(s)
- Shan Hui
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Department of Geriatric Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yu Yang
- Department of Geriatric Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wei-jun Peng
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Chen-xia Sheng
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wei Gong
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Shuai Chen
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Pan-pan Xu
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhe Wang
- Department of Integrated Chinese and Western Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
85
|
Datta A, Chai YL, Tan JM, Lee JH, Francis PT, Chen CP, Sze SK, Lai MKP. An iTRAQ-based proteomic analysis reveals dysregulation of neocortical synaptopodin in Lewy body dementias. Mol Brain 2017; 10:36. [PMID: 28800743 PMCID: PMC5553757 DOI: 10.1186/s13041-017-0316-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/21/2017] [Indexed: 02/08/2023] Open
Abstract
Lewy body dementias are the second most common cause of neurodegenerative dementia in the elderly after Alzheimer's disease (AD). The two clinical subgroups of Lewy body dementias, namely, dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD), are differentiated by the chronology of cognitive symptoms relative to parkinsonism. At present, there remains a debate on whether DLB and PDD are separate disease entities, or fall within the same spectrum of Lewy body dementias. In this study, we compared the detergent-soluble proteome via an 8-plex isobaric tag for relative and absolute quantitation (iTRAQ) analysis of pooled lysates from the prefrontal cortex (BA9) of DLB (n = 19) and PDD (n = 21) patients matched a priori for amyloid (total Aβ42) burden, semi-quantitative scores for Lewy bodies and neurofibrillary tangles together with age-matched control (n = 21) subjects. A total of 1914 proteins were confidently identified by iTRAQ (false discovery rate = 0%). None of the proteins showed a significant yet opposite regulation in between DLB and PDD when compared to aged controls in the proteomic data set as well as following immunoblot analysis of the pooled and individual lysates involving all 61 subjects. The postsynaptic protein, synaptopodin (SYNPO) was significantly down-regulated in both DLB and PDD subgroups, suggesting a defective synaptic transmission in the demented patients. In conclusion, the largely similar proteome of DLB and PDD matched for amyloid burden suggests that variations in concomitant AD-related pathology, abnormal post-translational modifications or protein-protein interactions, defective intracellular trafficking or misfolding of proteins could play a part in driving the clinically observed differences between these two subgroups of Lewy body dementias. This further indicates that amyloid-targeting therapeutic strategies may show different efficacies in DLB versus PDD.
Collapse
Affiliation(s)
- Arnab Datta
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore. .,Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, Building 10 Room 6N318, Bethesda, MD 20814, USA.
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore
| | - Jing Min Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore
| | - Jasinda H Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore
| | - Paul T Francis
- Wolfson Centre for Age-related Diseases, King's College London, Guy's Campus, St Thomas Street, London SE1 1UL, UK
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore. .,Wolfson Centre for Age-related Diseases, King's College London, Guy's Campus, St Thomas Street, London SE1 1UL, UK.
| |
Collapse
|
86
|
Bao W, Jia H, Finnema S, Cai Z, Carson RE, Huang YH. PET Imaging for Early Detection of Alzheimer's Disease: From Pathologic to Physiologic Biomarkers. PET Clin 2017; 12:329-350. [PMID: 28576171 DOI: 10.1016/j.cpet.2017.03.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This article describes the application of various PET imaging agents in the investigation and diagnosis of Alzheimer's disease (AD), including radiotracers for pathologic biomarkers of AD such as β-amyloid deposits and tau protein aggregates, and the neuroinflammation biomarker 18 kDa translocator protein, as well as physiologic biomarkers, such as cholinergic receptors, glucose metabolism, and the synaptic density biomarker synaptic vesicle glycoprotein 2A. Potential of these biomarkers for early AD diagnosis is also assessed.
Collapse
Affiliation(s)
- Weiqi Bao
- PET Center, Huanshan Hospital, Fudan University, No. 518, East Wuzhong Road, Xuhui District, Shanghai 200235, China
| | - Hongmei Jia
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, No. 19, Xinjiekouwai Street, Beijing 10075, China
| | - Sjoerd Finnema
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Yiyun Henry Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA.
| |
Collapse
|
87
|
Pathological Role of Peptidyl-Prolyl Isomerase Pin1 in the Disruption of Synaptic Plasticity in Alzheimer's Disease. Neural Plast 2017; 2017:3270725. [PMID: 28458925 PMCID: PMC5385220 DOI: 10.1155/2017/3270725] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 12/12/2016] [Indexed: 01/18/2023] Open
Abstract
Synaptic loss is the structural basis for memory impairment in Alzheimer's disease (AD). While the underlying pathological mechanism remains elusive, it is known that misfolded proteins accumulate as β-amyloid (Aβ) plaques and hyperphosphorylated Tau tangles decades before the onset of clinical disease. The loss of Pin1 facilitates the formation of these misfolded proteins in AD. Pin1 protein controls cell-cycle progression and determines the fate of proteins by the ubiquitin proteasome system. The activity of the ubiquitin proteasome system directly affects the functional and structural plasticity of the synapse. We localized Pin1 to dendritic rafts and postsynaptic density (PSD) and found the pathological loss of Pin1 within the synapses of AD brain cortical tissues. The loss of Pin1 activity may alter the ubiquitin-regulated modification of PSD proteins and decrease levels of Shank protein, resulting in aberrant synaptic structure. The loss of Pin1 activity, induced by oxidative stress, may also render neurons more susceptible to the toxicity of oligomers of Aβ and to excitation, thereby inhibiting NMDA receptor-mediated synaptic plasticity and exacerbating NMDA receptor-mediated synaptic degeneration. These results suggest that loss of Pin1 activity could lead to the loss of synaptic plasticity in the development of AD.
Collapse
|
88
|
Emerging Synaptic Molecules as Candidates in the Etiology of Neurological Disorders. Neural Plast 2017; 2017:8081758. [PMID: 28331639 PMCID: PMC5346360 DOI: 10.1155/2017/8081758] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023] Open
Abstract
Synapses are complex structures that allow communication between neurons in the central nervous system. Studies conducted in vertebrate and invertebrate models have contributed to the knowledge of the function of synaptic proteins. The functional synapse requires numerous protein complexes with specialized functions that are regulated in space and time to allow synaptic plasticity. However, their interplay during neuronal development, learning, and memory is poorly understood. Accumulating evidence links synapse proteins to neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. In this review, we describe the way in which several proteins that participate in cell adhesion, scaffolding, exocytosis, and neurotransmitter reception from presynaptic and postsynaptic compartments, mainly from excitatory synapses, have been associated with several synaptopathies, and we relate their functions to the disease phenotype.
Collapse
|
89
|
Ji XF, Chi TY, Liu P, Li LY, Xu JK, Xu Q, Zou LB, Meng DL. The total triterpenoid saponins of Xanthoceras sorbifolia improve learning and memory impairments through against oxidative stress and synaptic damage. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 25:15-24. [PMID: 28190466 DOI: 10.1016/j.phymed.2016.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 12/02/2016] [Accepted: 12/15/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND X. sorbifolia is a widely cultivated ecologicalcrop in the north of China which is used to produce biodiesel fuel. It also possesses special medicinal value and has attracted keen interests of researchers to explore its bioactivity. PURPOSE To extract the total triterpenoid saponins from the husk of X. sorbifolia (TSX) and investigate its effects on Alzheimer's disease (AD). STUDY DESIGN TSX was prepared via modern extraction techniques. Its effects on two AD animal models, as well as the preliminary mechanism were investigated comprehensively. METHODS The behavioral experiments including Y maze test, Morris water maze test and passive avoidance test were performed to observe the learning and memory abilities of the animals. ELISA assays, transmission electron microscope observation and Western blotting were employed in mechanism study. RESULTS TSX, the main composition of X. sorbifolia, accounted for 88.77% in the plant material. It could significantly increase the spontaneous alternation in Y maze test (F (6, 65)=3.209, P<0.01), prolong the swimming time in the fourth quadrant in probe test of Morris water maze test (F (6, 71)=4.019, P<0.01), and increase the escape latency in passive avoidance test (F (6, 65)=3.684, P<0.01) in AD model animals. The preliminary mechanism research revealed that TSX could significantly increase the contents of hippocampal Ach and ChAT, and enhance activity of ChAT in hippocampus of quinolinic acid injected rats (F (5, 61)=3.915, P 0.01; F (5, 61)=3.623, P<0.01, F (5, 61)=4.344, P<0.01, respectively). It could also increase the activities of T-AOC and T-SOD, and decrease the content of MDA in hippocampus of Aβ1-42 injected mice (F (5, 30)=5.193, P<0.01, F (5, 30)=2.865, P<0.05, F (5, 30)=4.735, P<0.01, respectively). Moreover, it significantly increased the expressions of SYP, PSD-95 and GAP-43 in hippocampus (F (4, 27)=3.495, P<0.05; F (4, 27)=2.965, P<0.05; F (4, 27)=4.365, P<0.01, respectively), and improved the synaptic ultra-structure damage in model rats. CONCLUSION TSX could significantly improve the impairments of learning and memory. The preliminary mechanism might associate with its protection effects against oxidative stress damage, cholinergic system deficiency and synaptic damage. TSX are perfectly suitable for AD patients as medicine or functional food, which would be a new candidate to treat AD.
Collapse
Affiliation(s)
- Xue-Fei Ji
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103 Wenhua road, Shenyang 110016, China.
| | - Tian-Yan Chi
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103 Wenhua road, Shenyang 110016, China
| | - Peng Liu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103 Wenhua road, Shenyang 110016, China
| | - Lu-Yi Li
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103 Wenhua road, Shenyang 110016, China
| | - Ji-Kai Xu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103 Wenhua road, Shenyang 110016, China
| | - Qian Xu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103 Wenhua road, Shenyang 110016, China
| | - Li-Bo Zou
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, No.103 Wenhua road, Shenyang 110016, China.
| | - Da-Li Meng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, No.103 Wenhua road, Shenyang 110016, China.
| |
Collapse
|
90
|
Orr AA, Wördehoff MM, Hoyer W, Tamamis P. Uncovering the Binding and Specificity of β-Wrapins for Amyloid-β and α-Synuclein. J Phys Chem B 2016; 120:12781-12794. [DOI: 10.1021/acs.jpcb.6b08485] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Asuka A. Orr
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
| | - Michael M. Wördehoff
- Institut
für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40204 Düsseldorf, Germany
| | - Wolfgang Hoyer
- Institut
für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40204 Düsseldorf, Germany
- Institute
of Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| | - Phanourios Tamamis
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
| |
Collapse
|
91
|
Soluble Amyloid-beta Aggregates from Human Alzheimer's Disease Brains. Sci Rep 2016; 6:38187. [PMID: 27917876 PMCID: PMC5137165 DOI: 10.1038/srep38187] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/04/2016] [Indexed: 11/09/2022] Open
Abstract
Soluble amyloid-beta (Aβ) aggregates likely contribute substantially to the dementia that characterizes Alzheimer's disease. However, despite intensive study of in vitro preparations and animal models, little is known about the characteristics of soluble Aβ aggregates in the human Alzheimer's disease brain. Here we present a new method for extracting soluble Aβ aggregates from human brains, separating them from insoluble aggregates and Aβ monomers using differential ultracentrifugation, and purifying them >6000 fold by dual antibody immunoprecipitation. The method resulted in <40% loss of starting material, no detectible ex vivo aggregation of monomeric Aβ, and no apparent ex vivo alterations in soluble aggregate sizes. By immunoelectron microscopy, soluble Aβ aggregates typically appear as clusters of 10-20 nanometer diameter ovoid structures with 2-3 amino-terminal Aβ antibody binding sites, distinct from previously characterized structures. This approach may facilitate investigation into the characteristics of native soluble Aβ aggregates, and deepen our understanding of Alzheimer's dementia.
Collapse
|
92
|
Mufson EJ, Ikonomovic MD, Counts SE, Perez SE, Malek-Ahmadi M, Scheff SW, Ginsberg SD. Molecular and cellular pathophysiology of preclinical Alzheimer's disease. Behav Brain Res 2016; 311:54-69. [PMID: 27185734 PMCID: PMC4931948 DOI: 10.1016/j.bbr.2016.05.030] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/12/2016] [Accepted: 05/12/2016] [Indexed: 12/19/2022]
Abstract
Although the two pathological hallmarks of Alzheimer's disease (AD), senile plaques composed of amyloid-β (Aβ) peptides and neurofibrillary tangles (NFTs) consisting of hyperphosphorylated tau, have been studied extensively in postmortem AD and relevant animal and cellular models, the pathogenesis of AD remains unknown, particularly in the early stages of the disease where therapies presumably would be most effective. We and others have demonstrated that Aβ plaques and NFTs are present in varying degrees before the onset and throughout the progression of dementia. In this regard, aged people with no cognitive impairment (NCI), mild cognitive impairment (MCI, a presumed prodromal AD transitional state, and AD all present at autopsy with varying levels of pathological hallmarks. Cognitive decline, a requisite for the clinical diagnosis of dementia associated with AD, generally correlates better with NFTs than Aβ plaques. However, correlations are even higher between cognitive decline and synaptic loss. In this review, we illustrate relevant clinical pathological research in preclinical AD and throughout the progression of dementia in several areas including Aβ and tau pathobiology, single population expression profiling of vulnerable hippocampal and basal forebrain neurons, neuroplasticity, neuroimaging, cerebrospinal fluid (CSF) biomarker studies and their correlation with antemortem cognitive endpoints. In each of these areas, we provide evidence for the importance of studying the pathological hallmarks of AD not in isolation, but rather in conjunction with other molecular, cellular, and imaging markers to provide a more systematic and comprehensive assessment of the multiple changes that occur during the transition from NCI to MCI to frank AD.
Collapse
Affiliation(s)
- Elliott J Mufson
- Departments of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, United States.
| | - Milos D Ikonomovic
- Departments of Neurology and Psychiatry, University of Pittsburgh, and Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine, Department of Family Medicine, Hauenstien Neuroscience Institute, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, United States
| | - Sylvia E Perez
- Departments of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| | | | - Stephen W Scheff
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Department of Psychiatry, Department of Neuroscience & Physiology, New York University Langone Medical Center, Orangeburg, NY, United States
| |
Collapse
|
93
|
Lepeta K, Lourenco MV, Schweitzer BC, Martino Adami PV, Banerjee P, Catuara-Solarz S, de La Fuente Revenga M, Guillem AM, Haidar M, Ijomone OM, Nadorp B, Qi L, Perera ND, Refsgaard LK, Reid KM, Sabbar M, Sahoo A, Schaefer N, Sheean RK, Suska A, Verma R, Vicidomini C, Wright D, Zhang XD, Seidenbecher C. Synaptopathies: synaptic dysfunction in neurological disorders - A review from students to students. J Neurochem 2016; 138:785-805. [PMID: 27333343 PMCID: PMC5095804 DOI: 10.1111/jnc.13713] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/03/2016] [Accepted: 06/06/2016] [Indexed: 12/12/2022]
Abstract
Synapses are essential components of neurons and allow information to travel coordinately throughout the nervous system to adjust behavior to environmental stimuli and to control body functions, memories, and emotions. Thus, optimal synaptic communication is required for proper brain physiology, and slight perturbations of synapse function can lead to brain disorders. In fact, increasing evidence has demonstrated the relevance of synapse dysfunction as a major determinant of many neurological diseases. This notion has led to the concept of synaptopathies as brain diseases with synapse defects as shared pathogenic features. In this review, which was initiated at the 13th International Society for Neurochemistry Advanced School, we discuss basic concepts of synapse structure and function, and provide a critical view of how aberrant synapse physiology may contribute to neurodevelopmental disorders (autism, Down syndrome, startle disease, and epilepsy) as well as neurodegenerative disorders (Alzheimer and Parkinson disease). We finally discuss the appropriateness and potential implications of gathering synapse diseases under a single term. Understanding common causes and intrinsic differences in disease-associated synaptic dysfunction could offer novel clues toward synapse-based therapeutic intervention for neurological and neuropsychiatric disorders. In this Review, which was initiated at the 13th International Society for Neurochemistry (ISN) Advanced School, we discuss basic concepts of synapse structure and function, and provide a critical view of how aberrant synapse physiology may contribute to neurodevelopmental (autism, Down syndrome, startle disease, and epilepsy) as well as neurodegenerative disorders (Alzheimer's and Parkinson's diseases), gathered together under the term of synaptopathies. Read the Editorial Highlight for this article on page 783.
Collapse
Affiliation(s)
- Katarzyna Lepeta
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Barbara C Schweitzer
- Department for Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
| | - Pamela V Martino Adami
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir-IIBBA-CONICET, Buenos Aires, Argentina
| | - Priyanjalee Banerjee
- Department of Biochemistry, Institute of Post Graduate Medical Education & Research, Kolkata, West Bengal, India
| | - Silvina Catuara-Solarz
- Systems Biology Program, Cellular and Systems Neurobiology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Mario de La Fuente Revenga
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Alain Marc Guillem
- Laboratorio de Neurotoxicología, Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México D.F. 07000, Mexico
| | - Mouna Haidar
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Omamuyovwi M Ijomone
- Department of Human Anatomy, Cross River University of Technology, Okuku Campus, Cross River, Nigeria
| | - Bettina Nadorp
- The Department of Biological Chemistry, The Edmond and Lily Safra Center for Brain Sciences, The Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Israel
| | - Lin Qi
- Laboratory of Molecular Neuro-Oncology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, United States of America
| | - Nirma D Perera
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Louise K Refsgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Kimberley M Reid
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Mariam Sabbar
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Arghyadip Sahoo
- Department of Biochemistry, Midnapore Medical College, West Bengal University of Health Sciences, West Bengal, India
| | - Natascha Schaefer
- Institute for Clinical Neurobiology, Julius-Maximilians-University of Wuerzburg, Wuerzburg, Germany
| | - Rebecca K Sheean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Anna Suska
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Rajkumar Verma
- Department of Neurosciences Uconn Health Center, Farmington, CT, United States of America
| | | | - Dean Wright
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Xing-Ding Zhang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Constanze Seidenbecher
- Department for Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany. .,Center for Behavioral Brain Sciences (CBBS) Magdeburg, Magdeburg, Germany.
| |
Collapse
|
94
|
Decock M, Stanga S, Octave JN, Dewachter I, Smith SO, Constantinescu SN, Kienlen-Campard P. Glycines from the APP GXXXG/GXXXA Transmembrane Motifs Promote Formation of Pathogenic Aβ Oligomers in Cells. Front Aging Neurosci 2016; 8:107. [PMID: 27242518 PMCID: PMC4861705 DOI: 10.3389/fnagi.2016.00107] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/25/2016] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder characterized by progressive cognitive decline leading to dementia. The amyloid precursor protein (APP) is a ubiquitous type I transmembrane (TM) protein sequentially processed to generate the β-amyloid peptide (Aβ), the major constituent of senile plaques that are typical AD lesions. There is a growing body of evidence that soluble Aβ oligomers correlate with clinical symptoms associated with the disease. The Aβ sequence begins in the extracellular juxtamembrane region of APP and includes roughly half of the TM domain. This region contains GXXXG and GXXXA motifs, which are critical for both TM protein interactions and fibrillogenic properties of peptides derived from TM α-helices. Glycine-to-leucine mutations of these motifs were previously shown to affect APP processing and Aβ production in cells. However, the detailed contribution of these motifs to APP dimerization, their relation to processing, and the conformational changes they can induce within Aβ species remains undefined. Here, we describe highly resistant Aβ42 oligomers that are produced in cellular membrane compartments. They are formed in cells by processing of the APP amyloidogenic C-terminal fragment (C99), or by direct expression of a peptide corresponding to Aβ42, but not to Aβ40. By a point-mutation approach, we demonstrate that glycine-to-leucine mutations in the G29XXXG33 and G38XXXA42 motifs dramatically affect the Aβ oligomerization process. G33 and G38 in these motifs are specifically involved in Aβ oligomerization; the G33L mutation strongly promotes oligomerization, while G38L blocks it with a dominant effect on G33 residue modification. Finally, we report that the secreted Aβ42 oligomers display pathological properties consistent with their suggested role in AD, but do not induce toxicity in survival assays with neuronal cells. Exposure of neurons to these Aβ42 oligomers dramatically affects neuronal differentiation and, consequently, neuronal network maturation.
Collapse
Affiliation(s)
- Marie Decock
- CEMO-Alzheimer Dementia, Institute of Neuroscience, Université Catholique de Louvain Brussels, Belgium
| | - Serena Stanga
- CEMO-Alzheimer Dementia, Institute of Neuroscience, Université Catholique de Louvain Brussels, Belgium
| | - Jean-Noël Octave
- CEMO-Alzheimer Dementia, Institute of Neuroscience, Université Catholique de Louvain Brussels, Belgium
| | - Ilse Dewachter
- CEMO-Alzheimer Dementia, Institute of Neuroscience, Université Catholique de Louvain Brussels, Belgium
| | - Steven O Smith
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook NY, USA
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research - de Duve Institute, Université Catholique de Louvain Brussels, Belgium
| | - Pascal Kienlen-Campard
- CEMO-Alzheimer Dementia, Institute of Neuroscience, Université Catholique de Louvain Brussels, Belgium
| |
Collapse
|
95
|
Synaptic Cell Adhesion Molecules in Alzheimer's Disease. Neural Plast 2016; 2016:6427537. [PMID: 27242933 PMCID: PMC4868906 DOI: 10.1155/2016/6427537] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/13/2016] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative brain disorder associated with the loss of synapses between neurons in the brain. Synaptic cell adhesion molecules are cell surface glycoproteins which are expressed at the synaptic plasma membranes of neurons. These proteins play key roles in formation and maintenance of synapses and regulation of synaptic plasticity. Genetic studies and biochemical analysis of the human brain tissue, cerebrospinal fluid, and sera from AD patients indicate that levels and function of synaptic cell adhesion molecules are affected in AD. Synaptic cell adhesion molecules interact with Aβ, a peptide accumulating in AD brains, which affects their expression and synaptic localization. Synaptic cell adhesion molecules also regulate the production of Aβ via interaction with the key enzymes involved in Aβ formation. Aβ-dependent changes in synaptic adhesion affect the function and integrity of synapses suggesting that alterations in synaptic adhesion play key roles in the disruption of neuronal networks in AD.
Collapse
|
96
|
He Y, Wang P, Wei P, Feng H, Ren Y, Yang J, Rao Y, Shi J, Tian J. Effects of curcumin on synapses in APPswe/PS1dE9 mice. Int J Immunopathol Pharmacol 2016; 29:217-25. [PMID: 26957323 DOI: 10.1177/0394632016638099] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 02/16/2016] [Indexed: 12/13/2022] Open
Abstract
Significant losses of synapses have been demonstrated in studies of Alzheimer's disease (AD), but structural and functional changes in synapses that depend on alterations of the postsynaptic density (PSD) area occur prior to synaptic loss and play a crucial role in the pathology of AD. Evidence suggests that curcumin can ameliorate the learning and memory deficits of AD. To investigate the effects of curcumin on synapses, APPswe/PS1dE9 double transgenic mice (an AD model) were used, and the ultra-structures of synapses and synapse-associated proteins were observed. Six months after administration, few abnormal synapses were observed upon electron microscopy in the hippocampal CA1 areas of the APPswe/PS1dE9 double transgenic mice. The treatment of the mice with curcumin resulted in improvements in the quantity and structure of the synapses. Immunohistochemistry and western blot analyses revealed that the expressions of PSD95 and Shank1 were reduced in the hippocampal CA1 areas of the APPswe/PS1dE9 double transgenic mice, but curcumin treatment increased the expressions of these proteins. Our findings suggest that curcumin improved the structure and function of the synapses by regulating the synapse-related proteins PSD95 and Shank1.
Collapse
Affiliation(s)
- Yingkun He
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, PR China
| | - Pengwen Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, PR China
| | - Peng Wei
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Jiaozuo Hospital of Traditional Chinese Medicine, Jiaozuo City, Henan Province, PR China
| | - Huili Feng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, PR China
| | - Ying Ren
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, PR China
| | - Jinduo Yang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, PR China
| | - Yingxue Rao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China University of Washington, Seattle, WA, USA
| | - Jing Shi
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Beijing University of Chinese Medicine, BUCM Neurology Center, Dongzhimen Hospital, Beijing, PR China
| | - Jinzhou Tian
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Beijing University of Chinese Medicine, BUCM Neurology Center, Dongzhimen Hospital, Beijing, PR China
| |
Collapse
|
97
|
AC-3933, a benzodiazepine partial inverse agonist, improves memory performance in MK-801-induced amnesia mouse model. Pharmacol Biochem Behav 2016; 144:45-52. [PMID: 26946254 DOI: 10.1016/j.pbb.2016.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/06/2016] [Accepted: 03/01/2016] [Indexed: 01/28/2023]
Abstract
AC-3933, a novel benzodiazepine receptor partial inverse agonist, is a drug candidate for cognitive disorders including Alzheimer's disease. We have previously reported that AC-3933 enhances acetylcholine release in the rat hippocampus and ameliorates scopolamine-induced memory impairment and age-related cognitive decline in both rats and mice. In this study, we further evaluated the procognitive effect of AC-3933 on memory impairment induced by MK-801, an N-methyl-d-aspartate receptor antagonist, in mice. Unlike the acetylcholinesterase inhibitor donepezil and the benzodiazepine receptor inverse agonist FG-7142, oral administration of AC-3933 significantly ameliorated MK-801-induced memory impairment in the Y-maze test and in the object location test. Interestingly, the procognitive effects of AC-3933 on MK-801-induced memory impairment were not affected by the benzodiazepine receptor antagonist flumazenil, although this was not the case for the beneficial effects of AC-3933 on scopolamine-induced memory deficit. Moreover, the onset of AC-3933 ameliorating effect on scopolamine- or MK-801-induced memory impairment was different in the Y-maze test. Taken together, these results indicate that AC-3933 improves memory deficits caused by both cholinergic and glutamatergic hypofunction and suggest that the ameliorating effect of AC-3933 on MK-801-induced memory impairment is mediated by a mechanism other than inverse activation of the benzodiazepine receptor.
Collapse
|
98
|
ElHajj Z, Cachot A, Müller T, Riederer IM, Riederer BM. Effects of postmortem delays on protein composition and oxidation. Brain Res Bull 2016; 121:98-104. [DOI: 10.1016/j.brainresbull.2016.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 01/05/2016] [Accepted: 01/06/2016] [Indexed: 10/22/2022]
|
99
|
Posttranslational Modifications Regulate the Postsynaptic Localization of PSD-95. Mol Neurobiol 2016; 54:1759-1776. [PMID: 26884267 DOI: 10.1007/s12035-016-9745-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/22/2016] [Indexed: 01/08/2023]
Abstract
The postsynaptic density (PSD) consists of a lattice-like array of interacting proteins that organizes and stabilizes synaptic receptors, ion channels, structural proteins, and signaling molecules required for normal synaptic transmission and synaptic function. The scaffolding and hub protein postsynaptic density protein-95 (PSD-95) is a major element of central chemical synapses and interacts with glutamate receptors, cell adhesion molecules, and cytoskeletal elements. In fact, PSD-95 can regulate basal synaptic stability as well as the activity-dependent structural plasticity of the PSD and, therefore, of the excitatory chemical synapse. Several studies have shown that PSD-95 is highly enriched at excitatory synapses and have identified multiple protein structural domains and protein-protein interactions that mediate PSD-95 function and trafficking to the postsynaptic region. PSD-95 is also a target of several signaling pathways that induce posttranslational modifications, including palmitoylation, phosphorylation, ubiquitination, nitrosylation, and neddylation; these modifications determine the synaptic stability and function of PSD-95 and thus regulate the fates of individual dendritic spines in the nervous system. In the present work, we review the posttranslational modifications that regulate the synaptic localization of PSD-95 and describe their functional consequences. We also explore the signaling pathways that induce such changes.
Collapse
|
100
|
de Wilde MC, Overk CR, Sijben JW, Masliah E. Meta-analysis of synaptic pathology in Alzheimer's disease reveals selective molecular vesicular machinery vulnerability. Alzheimers Dement 2016; 12:633-44. [PMID: 26776762 DOI: 10.1016/j.jalz.2015.12.005] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/02/2015] [Accepted: 12/04/2015] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Loss of synapses best correlates to cognitive deficits in Alzheimer's disease (AD) in which oligomeric neurotoxic species of amyloid-β appears to contribute synaptic pathology. Although a number of clinical pathologic studies have been performed with limited sample size, there are no systematic studies encompassing large samples. Therefore, we performed a meta-analysis study. METHODS We identified 417 publications reporting postmortem synapse and synaptic marker loss from AD patients. Two meta-analyses were performed using a single database of subselected publications and calculating the standard mean differences. RESULTS Meta-analysis confirmed synaptic loss in selected brain regions is an early event in AD pathogenesis. The second meta-analysis of 57 synaptic markers revealed that presynaptic makers were affected more than postsynaptic markers. DISCUSSION The present meta-analysis study showed a consistent synaptic loss across brain regions and that molecular machinery including endosomal pathways, vesicular assembly mechanisms, glutamate receptors, and axonal transport are often affected.
Collapse
Affiliation(s)
- Martijn C de Wilde
- Nutricia Advanced Medical Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Cassia R Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - John W Sijben
- Nutricia Advanced Medical Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Department of Pathology, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|