101
|
Gallardo G, Holtzman DM. Amyloid-β and Tau at the Crossroads of Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:187-203. [PMID: 32096039 DOI: 10.1007/978-981-32-9358-8_16] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia characterized neuropathologically by senile plaques and neurofibrillary tangles (NFTs). Early breakthroughs in AD research led to the discovery of amyloid-β as the major component of senile plaques and tau protein as the major component of NFTs. Shortly following the identification of the amyloid-β (Aβ) peptide was the discovery that a genetic mutation in the amyloid precursor protein (APP), a type1 transmembrane protein, can be a cause of autosomal dominant familial AD (fAD). These discoveries, coupled with other breakthroughs in cell biology and human genetics, have led to a theory known as the "amyloid hypothesis", which postulates that amyloid-β is the predominant driving factor in AD development. Nonetheless, more recent advances in imaging analysis, biomarkers and mouse models are now redefining this original hypothesis, as it is likely amyloid-β, tau and other pathophysiological mechanism such as inflammation, come together at a crossroads that ultimately leads to the development of AD.
Collapse
Affiliation(s)
- Gilbert Gallardo
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA. .,Hope Center for Neurological Disorders, Washington University, St. Louis, MO, USA. .,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA.
| |
Collapse
|
102
|
De Araujo Herculano B, Wang Z, Song W. A Novel Cell-based β-secretase Enzymatic Assay for Alzheimer's Disease. Curr Alzheimer Res 2018; 16:128-134. [PMID: 30543168 DOI: 10.2174/1567205016666181212151540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/19/2018] [Accepted: 11/28/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Deposition of the amyloid β protein (Aβ) into neuritic plaques is the neuropathological hallmark of Alzheimer's Disease (AD). Aβ is generated through the cleavage of the Amyloid Precursor Protein (APP) by β-secretase and γ-secretase. Currently, the evaluation of APP cleavage by β-secretase in experimental settings has largely depended on models that do not replicate the physiological conditions of this process. OBJECTIVE To establish a novel live cell-based β-secretase enzymatic assay utilizing a novel chimeric protein that incorporates the natural sequence of APP and more closely replicates its cleavage by β-secretase under physiological conditions. METHODS We have developed a chimeric protein construct, ASGβ, incorporating the β-site cleavage sequence of APP targeted by β-secretase and its intracellular trafficking signal into a Phosphatase-eGFP secreted reporter system. Upon cleavage by β-secretase, ASGβ releases a phosphatase-containing portion that can be measured in the culture medium, and an intracellular fraction that can be detected through Western Blot. Subsequently, we have generated a cell line stably expressing ASGβ that can be utilized to assay β-secretase in real time. RESULTS ASGβ is specifically targeted by β-secretase, being cleaved exclusively at the site responsible for the generation of Aβ. Dosage response to β-secretase inhibitors shows that β-secretase activity can be positively correlated to phosphatase activity in culture media. CONCLUSION Our findings suggest this system could be a high-throughput tool to screen compounds that aim to modulate β-secretase activity and Aβ production under physiological conditions, as well as evaluating factors that regulate this cleavage.
Collapse
Affiliation(s)
- Bruno De Araujo Herculano
- Department of Psychiatry, Townsend Family Laboratories, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Zhe Wang
- Department of Psychiatry, Townsend Family Laboratories, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Weihong Song
- Department of Psychiatry, Townsend Family Laboratories, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
103
|
Zhang X, Fu Z, Meng L, He M, Zhang Z. The Early Events That Initiate β-Amyloid Aggregation in Alzheimer's Disease. Front Aging Neurosci 2018; 10:359. [PMID: 30542277 PMCID: PMC6277872 DOI: 10.3389/fnagi.2018.00359] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/22/2018] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by the development of amyloid plaques and neurofibrillary tangles (NFTs) consisting of aggregated β-amyloid (Aβ) and tau, respectively. The amyloid hypothesis has been the predominant framework for research in AD for over two decades. According to this hypothesis, the accumulation of Aβ in the brain is the primary factor initiating the pathogenesis of AD. However, it remains elusive what factors initiate Aβ aggregation. Studies demonstrate that AD has multiple causes, including genetic and environmental factors. Furthermore, genetic factors, many age-related events and pathological conditions such as diabetes, traumatic brain injury (TBI) and aberrant microbiota also affect the aggregation of Aβ. Here we provide an overview of the age-related early events and other pathological processes that precede Aβ aggregation.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihui Fu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingyang He
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
104
|
Steiner H, Fukumori A, Tagami S, Okochi M. Making the final cut: pathogenic amyloid-β peptide generation by γ-secretase. Cell Stress 2018; 2:292-310. [PMID: 31225454 PMCID: PMC6551803 DOI: 10.15698/cst2018.11.162] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Alzheimer´s disease (AD) is a devastating neurodegenerative disease of the elderly population. Genetic evidence strongly suggests that aberrant generation and/or clearance of the neurotoxic amyloid-β peptide (Aβ) is triggering the disease. Aβ is generated from the amyloid precursor protein (APP) by the sequential cleavages of β- and γ-secretase. The latter cleavage by γ-secretase, a unique and fascinating four-component protease complex, occurs in the APP transmembrane domain thereby releasing Aβ species of 37-43 amino acids in length including the longer, highly pathogenic peptides Aβ42 and Aβ43. The lack of a precise understanding of Aβ generation as well as of the functions of other γ-secretase substrates has been one factor underlying the disappointing failure of γ-secretase inhibitors in clinical trials, but on the other side also been a major driving force for structural and in depth mechanistic studies on this key AD drug target in the past few years. Here we review recent breakthroughs in our understanding of how the γ-secretase complex recognizes substrates, of how it binds and processes β-secretase cleaved APP into different Aβ species, as well as the progress made on a question of outstanding interest, namely how clinical AD mutations in the catalytic subunit presenilin and the γ-secretase cleavage region of APP lead to relative increases of Aβ42/43. Finally, we discuss how the knowledge emerging from these studies could be used to therapeutically target this enzyme in a safe way.
Collapse
Affiliation(s)
- Harald Steiner
- Biomedical Center (BMC), Metabolic Biochemistry, Ludwig-Maximilians-University Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Akio Fukumori
- Department of Aging Neurobiology, National Center for Geriatrics and Gerontology, Obu & Department of Mental Health Promotion, Osaka University Graduate School of Medicine, Toyonaka, Japan
| | - Shinji Tagami
- Neuropsychiatry, Department of Integrated Medicine, Division of Internal Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masayasu Okochi
- Neuropsychiatry, Department of Integrated Medicine, Division of Internal Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
105
|
Xhima K, Nabbouh F, Hynynen K, Aubert I, Tandon A. Noninvasive delivery of an α-synuclein gene silencing vector with magnetic resonance-guided focused ultrasound. Mov Disord 2018; 33:1567-1579. [PMID: 30264465 DOI: 10.1002/mds.101] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The characteristic progression of Lewy pathology in Parkinson's disease likely involves intercellular exchange and the accumulation of misfolded α-synuclein amplified by a prion-like self-templating mechanism. Silencing of the α-synuclein gene could provide long-lasting disease-modifying benefits by reducing the requisite substrate for the spreading aggregation. OBJECTIVES As a result of the poor penetration of viral vectors across the blood-brain barrier, gene therapy for central nervous system disorders requires direct injections into the affected brain regions, and invasiveness is further increased by the need for bilateral delivery to multiple brain regions. Here we test a noninvasive approach by combining low-intensity magnetic resonance-guided focused ultrasound and intravenous microbubbles that can transiently increase the access of brain impermeant therapeutic macromolecules to targeted brain regions. METHODS Transgenic mice expressing human α-synuclein were subjected to magnetic resonance-guided focused ultrasound targeted to 4 brain regions (hippocampus, substantia nigra, olfactory bulb, and dorsal motor nucleus) in tandem with intravenous microbubbles and an adeno-associated virus serotype 9 vector bearing a short hairpin RNA sequence targeting the α-synuclein gene. RESULTS One month following treatment, α-synuclein immunoreactivity was decreased in targeted brain regions, whereas other neuronal markers such as synaptophysin or tyrosine hydroxylase were unchanged, and cell death and glial activation remained at basal levels. CONCLUSIONS These results demonstrate that magnetic resonance-guided focused ultrasound can effectively, noninvasively, and simultaneously deliver viral vectors targeting α-synuclein to multiple brain areas. Importantly, this approach may be useful to alter the progression of Lewy pathology along selected neuronal pathways, particularly as prodromal PD markers improve early diagnoses. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Kristiana Xhima
- Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Fadl Nabbouh
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, Ontario, Canada
| | - Kullervo Hynynen
- Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Isabelle Aubert
- Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Anurag Tandon
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
106
|
Presenilin 1 deficiency suppresses autophagy in human neural stem cells through reducing γ-secretase-independent ERK/CREB signaling. Cell Death Dis 2018; 9:879. [PMID: 30158533 PMCID: PMC6115391 DOI: 10.1038/s41419-018-0945-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/18/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022]
Abstract
Autophagy impairment is commonly implicated in the pathological characteristic of Alzheimer’s disease (AD). Presenilin 1 (PS1) expression in human brain gradually decreases with age and its mutations account for the most common cases of early-onset familial Alzheimer’s disease (FAD). The dominant autophagy phenotypes occur in PS1-knockout and PS1 mutant neurons; it is still unknown whether PS1 deficiency causes serious autophagy impairment in neural stem cells (NSCs). Herein, we generated the heterozygote and homozygote of PS1 knockout in human induced pluripotent stem cells (iPSCs) via CRISPR/Cas9-based gene editing and differentiated them into human NSCs. In these human PS1-deficient NSCs, reduced autophagosome formation and downregulated expression of autophagy–lysosome pathway (ALP)-related mRNAs, as well as proteins were observed. Mechanistically, ERK/CREB inhibition and GSK3β activation had key roles in reducing TFEB expression in PS1-knockout NSCs. Pharmacological inhibition of GSK3β upregulated the expression of TFEB and ALP-related proteins in PS1-knockout NSCs, whereas this effect could be blocked by CREB inhibition. These findings demonstrate that PS1 deficiency causes autophagy suppression in human NSCs via downregulating ERK/CREB signaling.
Collapse
|
107
|
Paltsev MA, Zuev VA, Kozhevnikova EO, Linkova NS, Kvetnaia TV, Polyakova VO, Kvetnoy IM. Molecular Markers of Early Diagnosis of Alzheimer Disease: Prospects for Research in Peripheral Tissues. ADVANCES IN GERONTOLOGY 2018. [DOI: 10.1134/s2079057018020133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
108
|
Schilling S, Rahfeld JU, Lues I, Lemere CA. Passive Aβ Immunotherapy: Current Achievements and Future Perspectives. Molecules 2018; 23:molecules23051068. [PMID: 29751505 PMCID: PMC6099643 DOI: 10.3390/molecules23051068] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 12/28/2022] Open
Abstract
Passive immunotherapy has emerged as a very promising approach for the treatment of Alzheimer’s disease and other neurodegenerative disorders, which are characterized by the misfolding and deposition of amyloid peptides. On the basis of the amyloid hypothesis, the majority of antibodies in clinical development are directed against amyloid β (Aβ), the primary amyloid component in extracellular plaques. This review focuses on the current status of Aβ antibodies in clinical development, including their characteristics and challenges that came up in clinical trials with these new biological entities (NBEs). Emphasis is placed on the current view of common side effects observed with passive immunotherapy, so-called amyloid-related imaging abnormalities (ARIAs), and potential ways to overcome this issue. Among these new ideas, a special focus is placed on molecules that are directed against post-translationally modified variants of the Aβ peptide, an emerging approach for development of new antibody molecules.
Collapse
Affiliation(s)
- Stephan Schilling
- Fraunhofer Institute for Cell Therapy and Immunology, Department for Drug Design and Target Validation, 06120 Halle (Saale), Germany.
| | - Jens-Ulrich Rahfeld
- Fraunhofer Institute for Cell Therapy and Immunology, Department for Drug Design and Target Validation, 06120 Halle (Saale), Germany.
| | - Inge Lues
- Probiodrug AG, 06120 Halle (Saale), Germany.
| | - Cynthia A Lemere
- Ann Romney Center for Neurologic Diseases, Brigham and Womens's Hospital, Harvard Medical School, Boston, MA 02116, USA.
| |
Collapse
|
109
|
Beckert B, Acker-Palmer A, Volknandt W. Aβ42 oligomers impair the bioenergetic activity in hippocampal synaptosomes derived from APP-KO mice. Biol Chem 2018; 399:453-465. [PMID: 29337689 DOI: 10.1515/hsz-2017-0238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/20/2017] [Indexed: 11/15/2022]
Abstract
Employing hippocampal synaptosomes from amyloid precursor protein (APP)-deleted mice we analyzed the immediate effects of amyloid beta peptide 42 (Aβ42) peptide in its oligomeric or fibrillar assembly or of soluble amyloid precursor protein alpha (sAPPα) protein on their bioenergetic activity. Upon administration of oligomeric Aβ42 peptide for 30 min we observed a robust decrease both in mitochondrial activity and in mitochondrial membrane potential (MMP). In contrast the respective fibrillary or scrambled peptides showed no effect, indicating that inhibition strictly depends on the oligomerization status of the peptide. Hippocampal synaptosomes from old APP-KO mice revealed a further reduction of their already impaired bioenergetic activity upon incubation with 10 μm Aβ42 peptide. In addition we evaluated the influence of the sAPPα protein on mitochondrial activity of hippocampal synaptosomes derived from young or old APP-KO animals. In neither case 20 nm nor 200 nm sAPPα protein had an effect on mitochondrial metabolic activity. Our findings demonstrate that hippocampal synaptosomes derived from APP-KO mice are a most suitable model system to evaluate the impact of Aβ42 peptide on its bioenergetic activity and to further elucidate the molecular mechanisms underlying the impairments by oligomeric Aβ42 on mitochondrial function. Our data demonstrate that extracellular Aβ42 peptide is taken up into synaptosomes where it immediately attenuates mitochondrial activity.
Collapse
Affiliation(s)
- Benedikt Beckert
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt/Main, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt/Main, Germany
- Max Planck Institute for Brain Research, Max-von-Laue-Str. 4, D-60438 Frankfurt/Main, Germany
| | - Walter Volknandt
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, D-60438, Frankfurt/Main, Germany
- Department for Molecular and Cellular Neurobiology, Goethe University Frankfurt, Max-von-Laue-Str. 13, D-60438 Frankfurt, Germany
| |
Collapse
|
110
|
Galzitskaya OV, Galushko EI, Selivanova OM. Studies of the Process of Amyloid Formation by Aβ Peptide. BIOCHEMISTRY (MOSCOW) 2018; 83:S62-S80. [PMID: 29544432 DOI: 10.1134/s0006297918140079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Studies of the process of amyloid formation by Aβ peptide have been topical due to the critical role of this peptide in the pathogenesis of Alzheimer's disease. Many articles devoted to this process are available in the literature; however, none of them gives a detailed description of the mechanism of the process of generation of amyloids. Moreover, there are no reliable data on the influence of modified forms of Aβ peptide on its amyloid formation. To appreciate the role of Aβ aggregation in the pathogenesis of Alzheimer's disease and to develop a strategy for its treatment, it is necessary to have a well-defined description of the molecular mechanism underlying the formation of amyloids as well as the contribution of each intermediate to this process. We are convinced that a combined analysis of theoretical and experimental methods is a way for understanding molecular mechanisms of numerous diseases. Based on our experimental data and molecular modeling, we have constructed a general model of the process of amyloid formation by Aβ peptide. Using the data described in our previous publications, we propose a model of amyloid formation by this peptide that differs from the generally accepted model. Our model can be applied to other proteins and peptides as well. According to this model, the main building unit for the formation of amyloid fibrils is a ring-like oligomer. Upon interaction with each other, ring-like oligomers form long fibrils of different morphology. This mechanism of generation of amyloid fibrils may be common for other proteins and peptides.
Collapse
Affiliation(s)
- O V Galzitskaya
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | | | | |
Collapse
|
111
|
Wolfe MS. Dysfunctional γ-Secretase in Familial Alzheimer's Disease. Neurochem Res 2018; 44:5-11. [PMID: 29619615 PMCID: PMC6592691 DOI: 10.1007/s11064-018-2511-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 03/19/2018] [Indexed: 12/24/2022]
Abstract
Genetics strongly implicate the amyloid β-peptide (Aβ) in the pathogenesis of Alzheimer's disease. Dominant missense mutation in the presenilins and the amyloid precursor protein (APP) cause early-onset familial Alzheimer's disease (FAD). As presenilin is the catalytic component of the γ-secretase protease complex that produces Aβ from APP, mutation of the enzyme or substrate that produce Aβ leads to FAD. However, the mechanism by which presenilin mutations cause FAD has been controversial, with gain of function and loss of function offered as binary choices. This overview will instead present the case that presenilins are dysfunctional in FAD. γ-Secretase is a multi-functional enzyme that proteolyzes the APP transmembrane domain in a complex and processive manner. Reduction in a specific function-the carboxypeptidase trimming of initially formed long Aβ peptides containing most of the transmembrane domain to shorter secreted forms-is an emerging common feature of FAD-mutant γ-secretase complexes.
Collapse
Affiliation(s)
- Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, 66045, USA.
| |
Collapse
|
112
|
Lee M, Guo JP, Kennedy K, McGeer EG, McGeer PL. A Method for Diagnosing Alzheimer's Disease Based on Salivary Amyloid-β Protein 42 Levels. J Alzheimers Dis 2018; 55:1175-1182. [PMID: 27792013 DOI: 10.3233/jad-160748] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We have developed a non-invasive method of diagnosing Alzheimer's disease (AD), which can also predict the risk of its future onset. It is based on measuring salivary levels of amyloid-β protein terminating at position 42 (Aβ42). Brain deposits of this peptide are characteristic of AD. Biomarker studies indicate that such brain deposits commence a decade or more prior to clinical onset of the disease. We report here that Aβ42 is produced in all peripheral organs tested, thus establishing the generality of its production. We used this information to develop simple and sensitive tests to determine salivary Aβ42 levels. The levels were first stabilized by adding thioflavin S as an anti-aggregation agent and sodium azide as an anti-bacterial agent. We then quantitated the Aβ42 in a series of samples with ELISA type tests. Control cases showed almost identical levels of salivary Aβ42 regardless of sex or age. All AD cases secreted levels of Aβ42 more than double those of controls. Individuals at elevated risk of developing AD secreted levels comparable to the AD cases. The results establish that salivary Aβ42 levels can be used to diagnose AD as well as to predict the risk of its future onset.
Collapse
|
113
|
Jakobsen JE, Johansen MG, Schmidt M, Liu Y, Li R, Callesen H, Melnikova M, Habekost M, Matrone C, Bouter Y, Bayer TA, Nielsen AL, Duthie M, Fraser PE, Holm IE, Jørgensen AL. Expression of the Alzheimer's Disease Mutations AβPP695sw and PSEN1M146I in Double-Transgenic Göttingen Minipigs. J Alzheimers Dis 2018; 53:1617-30. [PMID: 27540966 DOI: 10.3233/jad-160408] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Mutations in the amyloid-β protein precursor gene (AβPP), the presenilin 1 gene (PSEN1) or the presenilin 2 gene (PSEN2) that increase production of the AβPP-derived peptide Aβ42 cause early-onset Alzheimer's disease. Rodent models of the disease show that further increase in Aβ42 production and earlier brain pathology can be obtained by coexpressing AβPP and PSEN1 mutations. To generate such elevated Aβ42 level in a large animal model, we produced Göttingen minipigs carrying in their genome one copy of a human PSEN1 cDNA with the Met146Ile (PSEN1M146I) mutation and three copies of a human AβPP695 cDNA with the Lys670Asn/Met671Leu (AβPPsw) double-mutation. Both transgenes were expressed in fibroblasts and in the brain, and their respective proteins were processed normally. Immunohistochemical staining with Aβ42-specific antibodies detected intraneuronal accumulation of Aβ42 in brains from a 10- and an 18-month-old pig. Such accumulation may represent an early event in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Jannik E Jakobsen
- Department of Biomedicine (East), Aarhus University, Aarhus C, Denmark
| | | | - Mette Schmidt
- Department of Large Animal Sciences/Reproduction, University of Copenhagen, Frederiksberg C, Denmark
| | - Ying Liu
- Department of Animal Science, Aarhus University, Tjele, Denmark
| | - Rong Li
- Department of Animal Science, Aarhus University, Tjele, Denmark
| | - Henrik Callesen
- Department of Animal Science, Aarhus University, Tjele, Denmark
| | | | - Mette Habekost
- Department of Biomedicine (East), Aarhus University, Aarhus C, Denmark
| | - Carmela Matrone
- Department of Biomedicine (East), Aarhus University, Aarhus C, Denmark
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medicine Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Thomas A Bayer
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medicine Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | | | - Monika Duthie
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Ida E Holm
- Department of Clinical Medicine, Aarhus University, Aarhus C, Denmark.,Department of Pathology, Randers Hospital, Randers, Denmark
| | | |
Collapse
|
114
|
Perleberg C, Kind A, Schnieke A. Genetically engineered pigs as models for human disease. Dis Model Mech 2018; 11:11/1/dmm030783. [PMID: 29419487 PMCID: PMC5818075 DOI: 10.1242/dmm.030783] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Genetically modified animals are vital for gaining a proper understanding of disease mechanisms. Mice have long been the mainstay of basic research into a wide variety of diseases but are not always the most suitable means of translating basic knowledge into clinical application. The shortcomings of rodent preclinical studies are widely recognised, and regulatory agencies around the world now require preclinical trial data from nonrodent species. Pigs are well suited to biomedical research, sharing many similarities with humans, including body size, anatomical features, physiology and pathophysiology, and they already play an important role in translational studies. This role is set to increase as advanced genetic techniques simplify the generation of pigs with precisely tailored modifications designed to replicate lesions responsible for human disease. This article provides an overview of the most promising and clinically relevant genetically modified porcine models of human disease for translational biomedical research, including cardiovascular diseases, cancers, diabetes mellitus, Alzheimer's disease, cystic fibrosis and Duchenne muscular dystrophy. We briefly summarise the technologies involved and consider the future impact of recent technical advances. Summary: An overview of porcine models of human disease, including cardiovascular diseases, cancers, diabetes mellitus, Alzheimer's disease, cystic fibrosis and Duchenne muscular dystrophy. We summarise the technologies involved and potential future impact of recent technical advances.
Collapse
Affiliation(s)
- Carolin Perleberg
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Alexander Kind
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| |
Collapse
|
115
|
Yener Ilce B, Cagin U, Yilmazer A. Cellular reprogramming: A new way to understand aging mechanisms. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 7. [PMID: 29350802 DOI: 10.1002/wdev.308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/29/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022]
Abstract
Increased life expectancy, due to the rise in life quality and the decline in mortality rates, is leading to a society in which the population aged 60 and over is growing more rapidly than the entire population. Although various models and model organisms have been employed to investigate the mechanism of aging, induced pluripotent stem cells (iPSCs) are useful candidates to study human aging and age-related human diseases. This work discusses how iPSCs can be used as an alternative to the model organisms such as yeast, Caenorhabditis elegans, Drosophila melanogaster, or the mouse. The main focus is the reprogramming technology of somatic cells which is thought to provide an important perspective for rejuvenation strategies. The effects and relationships between aging and cell reprogramming are discussed, and studies related to aging and cell reprogramming are critically reviewed. We believe that for future studies, different parameters and detailed quantitative experiments should be performed in order to clearly understand the effect of aging on human cell reprogramming with respect to programming efficiency and differentiation capacity. This way, new insights will be provided to prevent or even reverse the aging process. WIREs Dev Biol 2018, 7:e308. doi: 10.1002/wdev.308 This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cells and Aging Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cells and Disease.
Collapse
Affiliation(s)
| | | | - Acelya Yilmazer
- Biomedical Engineering Department, Engineering Faculty, Ankara University, Ankara, Turkey.,Stem Cell Institute, Ankara University, Ankara, Turkey
| |
Collapse
|
116
|
Fontana BD, Mezzomo NJ, Kalueff AV, Rosemberg DB. The developing utility of zebrafish models of neurological and neuropsychiatric disorders: A critical review. Exp Neurol 2018; 299:157-171. [DOI: 10.1016/j.expneurol.2017.10.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/15/2017] [Accepted: 10/04/2017] [Indexed: 12/30/2022]
|
117
|
Johnson DS, Li YM, Pettersson M, St George-Hyslop PH. Structural and Chemical Biology of Presenilin Complexes. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a024067. [PMID: 28320827 PMCID: PMC5710098 DOI: 10.1101/cshperspect.a024067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The presenilin proteins are the catalytic subunits of a tetrameric complex containing presenilin 1 or 2, anterior pharynx defective 1 (APH1), nicastrin, and PEN-2. Other components such as TMP21 may exist in a subset of specialized complexes. The presenilin complex is the founding member of a unique class of aspartyl proteases that catalyze the γ, ɛ, ζ site cleavage of the transmembrane domains of Type I membrane proteins including amyloid precursor protein (APP) and Notch. Here, we detail the structural and chemical biology of this unusual enzyme. Taken together, these studies suggest that the complex exists in several conformations, and subtle long-range (allosteric) shifts in the conformation of the complex underpin substrate access to the catalytic site and the mechanism of action for allosteric inhibitors and modulators. Understanding the mechanics of these shifts will facilitate the design of γ-secretase modulator (GSM) compounds that modulate the relative efficiency of γ, ɛ, ζ site cleavage and/or substrate specificity.
Collapse
Affiliation(s)
- Douglas S. Johnson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Martin Pettersson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | - Peter H. St George-Hyslop
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Cambridge CB2 0XY, United Kingdom,Tanz Centre for Research in Neurodegenerative Diseases and Departments of Medicine, Laboratory Medicine and Pathobiology, and Medical Biophysics, University of Toronto, Toronto, Ontario M5T 2S8, Canada
| |
Collapse
|
118
|
Guo J, Cheng J, North BJ, Wei W. Functional analyses of major cancer-related signaling pathways in Alzheimer's disease etiology. Biochim Biophys Acta Rev Cancer 2017; 1868:341-358. [PMID: 28694093 PMCID: PMC5675793 DOI: 10.1016/j.bbcan.2017.07.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disease and accounts for majority of human dementia. The hyper-phosphorylated tau-mediated intracellular neurofibrillary tangle and amyloid β-mediated extracellular senile plaque are characterized as major pathological lesions of AD. Different from the dysregulated growth control and ample genetic mutations associated with human cancers, AD displays damage and death of brain neurons in the absence of genomic alterations. Although various biological processes predominately governing tumorigenesis such as inflammation, metabolic alteration, oxidative stress and insulin resistance have been associated with AD genesis, the mechanistic connection of these biological processes and signaling pathways including mTOR, MAPK, SIRT, HIF, and the FOXO pathway controlling aging and the pathological lesions of AD are not well recapitulated. Hence, we performed a thorough review by summarizing the physiological roles of these key cancer-related signaling pathways in AD pathogenesis, comprising of the crosstalk of these pathways with neurofibrillary tangle and senile plaque formation to impact AD phenotypes. Importantly, the pharmaceutical investigations of anti-aging and AD relevant medications have also been highlighted. In summary, in this review, we discuss the potential role that cancer-related signaling pathways may play in governing the pathogenesis of AD, as well as their potential as future targeted strategies to delay or prevent aging-related diseases and combating AD.
Collapse
Affiliation(s)
- Jianping Guo
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ji Cheng
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Brian J North
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
119
|
Zhou R, Yang G, Shi Y. Dominant negative effect of the loss-of-function γ-secretase mutants on the wild-type enzyme through heterooligomerization. Proc Natl Acad Sci U S A 2017; 114:12731-12736. [PMID: 29078389 PMCID: PMC5715776 DOI: 10.1073/pnas.1713605114] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
γ-secretase is an intramembrane protease complex consisting of nicastrin, presenilin-1/2, APH-1a/b, and Pen-2. Hydrolysis of the 99-residue transmembrane fragment of amyloid precursor protein (APP-C99) by γ-secretase produces β-amyloid (Aβ) peptides. Pathogenic mutations in PSEN1 and PSEN2, which encode the catalytic subunit presenilin-1/2 of γ-secretase, lead to familial Alzheimer's disease in an autosomal dominant manner. However, the underlying mechanism of how the mutant PSEN gene may affect the function of the WT allele remains to be elucidated. Here we report that each of the loss-of-function γ-secretase variants that carries a PSEN1 mutation suppresses the protease activity of the WT γ-secretase on Aβ production. Each of these γ-secretase variants forms a stable oligomer with the WT γ-secretase in vitro in the presence of the detergent CHAPSO {3-[(3-cholamidopropyl)dimethylammonio]-2-hydroxy-1-propanesulfonate}, but not digitonin. Importantly, robust protease activity of γ-secretase is detectable in the presence of CHAPSO, but not digitonin. These experimental observations suggest a dominant negative effect of the γ-secretase, in which the protease activity of WT γ-secretase is suppressed by the loss-of-function γ-secretase variants through hetero-oligomerization. The relevance of this finding to the genesis of Alzheimer's disease is critically evaluated.
Collapse
Affiliation(s)
- Rui Zhou
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China;
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Guanghui Yang
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China;
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yigong Shi
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing 100084, China;
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Institute of Biology, Westlake Institute for Advanced Study, Xihu District, Hangzhou 310064, Zhejiang Province, China
| |
Collapse
|
120
|
Brody AH, Strittmatter SM. Synaptotoxic Signaling by Amyloid Beta Oligomers in Alzheimer's Disease Through Prion Protein and mGluR5. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:293-323. [PMID: 29413525 PMCID: PMC5835229 DOI: 10.1016/bs.apha.2017.09.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) represents an impending global health crisis, yet the complexity of AD pathophysiology has so far precluded the development of any interventions to successfully slow or halt AD progression. It is clear that accumulation of Amyloid-beta (Aβ) peptide triggers progressive synapse loss to cause AD symptoms. Once initiated by Aβ, disease progression is complicated and accelerated by inflammation and by tau pathology. The recognition that Aβ peptide assumes multiple distinct states and that soluble oligomeric species (Aβo) are critical for synaptic damage is central to molecular understanding of AD. This knowledge has led to the identification of specific Aβo receptors, such as cellular prion protein (PrPC), mediating synaptic toxicity and neuronal dysfunction. The identification of PrPC as an Aβo receptor has illuminated an Aβo-induced signaling cascade involving mGluR5, Fyn, and Pyk2 that links Aβ and tau pathologies. This pathway provides novel potential therapeutic targets for disease-modifying AD therapy. Here, we discuss the methods by which several putative Aβo receptors were identified. We also offer an in-depth examination of the known molecular mechanisms believed to mediate Aβo-induced synaptic dysfunction, toxicity, and memory dysfunction.
Collapse
Affiliation(s)
- A Harrison Brody
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT, United States; Yale University, New Haven, CT, United States
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT, United States; Yale University, New Haven, CT, United States.
| |
Collapse
|
121
|
Eskandari-Sedighi G, Daude N, Gapeshina H, Sanders DW, Kamali-Jamil R, Yang J, Shi B, Wille H, Ghetti B, Diamond MI, Janus C, Westaway D. The CNS in inbred transgenic models of 4-repeat Tauopathy develops consistent tau seeding capacity yet focal and diverse patterns of protein deposition. Mol Neurodegener 2017; 12:72. [PMID: 28978354 PMCID: PMC5628424 DOI: 10.1186/s13024-017-0215-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/27/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND MAPT mutations cause neurodegenerative diseases such as frontotemporal dementia but, strikingly, patients with the same mutation may have different clinical phenotypes. METHODS Given heterogeneities observed in a transgenic (Tg) mouse line expressing low levels of human (2 N, 4R) P301L Tau, we backcrossed founder stocks of mice to C57BL/6Tac, 129/SvEvTac and FVB/NJ inbred backgrounds to discern the role of genetic versus environmental effects on disease-related phenotypes. RESULTS Three inbred derivatives of a TgTauP301L founder line had similar quality and steady-state quantity of Tau production, accumulation of abnormally phosphorylated 64-68 kDa Tau species from 90 days of age onwards and neuronal loss in aged Tg mice. Variegation was not seen in the pattern of transgene expression and seeding properties in a fluorescence-based cellular assay indicated a single "strain" of misfolded Tau. However, in other regards, the aged Tg mice were heterogeneous; there was incomplete penetrance for Tau deposition despite maintained transgene expression in aged animals and, for animals with Tau deposits, distinctions were noted even within each subline. Three classes of rostral deposition in the cortex, hippocampus and striatum accounted for 75% of pathology-positive mice yet the mean ages of mice scored as class I, II or III were not significantly different and, hence, did not fit with a predictable progression from one class to another defined by chronological age. Two other patterns of Tau deposition designated as classes IV and V, occurred in caudal structures. Other pathology-positive Tg mice of similar age not falling within classes I-V presented with focal accumulations in additional caudal neuroanatomical areas including the locus coeruleus. Electron microscopy revealed that brains of Classes I, II and IV animals all exhibit straight filaments, but with coiled filaments and occasional twisted filaments apparent in Class I. Most strikingly, Class I, II and IV animals presented with distinct western blot signatures after trypsin digestion of sarkosyl-insoluble Tau. CONCLUSIONS Qualitative variations in the neuroanatomy of Tau deposition in genetically constrained slow models of primary Tauopathy establish that non-synchronous, focal events contribute to the pathogenic process. Phenotypic diversity in these models suggests a potential parallel to the phenotypic variation seen in P301L patients.
Collapse
Affiliation(s)
- Ghazaleh Eskandari-Sedighi
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Nathalie Daude
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada
| | - Hristina Gapeshina
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada
| | - David W Sanders
- Center for Alzheimer's and Neurodegenerative Diseases, UT Southwestern Medical Center, Dallas, USA
| | - Razieh Kamali-Jamil
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Jing Yang
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada
| | - Beipei Shi
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada
| | - Holger Wille
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, USA
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, UT Southwestern Medical Center, Dallas, USA
| | - Christopher Janus
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada. .,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
122
|
Chauhan R, Chen KF, Kent BA, Crowther DC. Central and peripheral circadian clocks and their role in Alzheimer's disease. Dis Model Mech 2017; 10:1187-1199. [PMID: 28993311 PMCID: PMC5665458 DOI: 10.1242/dmm.030627] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Molecular and cellular oscillations constitute an internal clock that tracks the time of day and permits organisms to optimize their behaviour and metabolism to suit the daily demands they face. The workings of this internal clock become impaired with age. In this review, we discuss whether such age-related impairments in the circadian clock interact with age-related neurodegenerative disorders, such as Alzheimer's disease. Findings from mouse and fly models of Alzheimer's disease have accelerated our understanding of the interaction between neurodegeneration and circadian biology. These models show that neurodegeneration likely impairs circadian rhythms either by damaging the central clock or by blocking its communication with other brain areas and with peripheral tissues. The consequent sleep and metabolic deficits could enhance the susceptibility of the brain to further degenerative processes. Thus, circadian dysfunction might be both a cause and an effect of neurodegeneration. We also discuss the primary role of light in the entrainment of the central clock and describe important, alternative time signals, such as food, that play a role in entraining central and peripheral circadian clocks. Finally, we propose how these recent insights could inform efforts to develop novel therapeutic approaches to re-entrain arrhythmic individuals with neurodegenerative disease.
Collapse
Affiliation(s)
- Ruchi Chauhan
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Ko-Fan Chen
- Institute of Neurology, UCL, London, WC1N 3BG, UK
| | - Brianne A Kent
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, V6T 1Z3, Canada
| | - Damian C Crowther
- Neuroscience, Innovative Medicines and Early Development, AstraZeneca, Granta Park, Cambridge, CB21 6GH, UK
| |
Collapse
|
123
|
Menzies FM, Fleming A, Caricasole A, Bento CF, Andrews SP, Ashkenazi A, Füllgrabe J, Jackson A, Jimenez Sanchez M, Karabiyik C, Licitra F, Lopez Ramirez A, Pavel M, Puri C, Renna M, Ricketts T, Schlotawa L, Vicinanza M, Won H, Zhu Y, Skidmore J, Rubinsztein DC. Autophagy and Neurodegeneration: Pathogenic Mechanisms and Therapeutic Opportunities. Neuron 2017; 93:1015-1034. [PMID: 28279350 DOI: 10.1016/j.neuron.2017.01.022] [Citation(s) in RCA: 839] [Impact Index Per Article: 104.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/11/2022]
Abstract
Autophagy is a conserved pathway that delivers cytoplasmic contents to the lysosome for degradation. Here we consider its roles in neuronal health and disease. We review evidence from mouse knockout studies demonstrating the normal functions of autophagy as a protective factor against neurodegeneration associated with intracytoplasmic aggregate-prone protein accumulation as well as other roles, including in neuronal stem cell differentiation. We then describe how autophagy may be affected in a range of neurodegenerative diseases. Finally, we describe how autophagy upregulation may be a therapeutic strategy in a wide range of neurodegenerative conditions and consider possible pathways and druggable targets that may be suitable for this objective.
Collapse
Affiliation(s)
- Fiona M Menzies
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Angeleen Fleming
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Andrea Caricasole
- Alzheimer's Research UK Cambridge Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK
| | - Carla F Bento
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Stephen P Andrews
- Alzheimer's Research UK Cambridge Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK
| | - Avraham Ashkenazi
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Jens Füllgrabe
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Anne Jackson
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Maria Jimenez Sanchez
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Cansu Karabiyik
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Floriana Licitra
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Ana Lopez Ramirez
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Mariana Pavel
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Claudia Puri
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Maurizio Renna
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Thomas Ricketts
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Lars Schlotawa
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Mariella Vicinanza
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Hyeran Won
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Ye Zhu
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - John Skidmore
- Alzheimer's Research UK Cambridge Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|
124
|
Genetics of Alzheimer's disease: From pathogenesis to clinical usage. J Clin Neurosci 2017; 45:1-8. [PMID: 28869135 DOI: 10.1016/j.jocn.2017.06.074] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/19/2017] [Indexed: 01/27/2023]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and has caused a major global health concern. Understanding the etiology of AD can be beneficial for the diagnosis and intervention of this disease. Genetics plays a vital role in the pathogenesis of AD. Research methods in genetics such as the linkage analysis, study of candidate genes, genome-wide association study (GWAS), and next-generation sequencing (NGS) technology help us map the genetic information in AD, which can not only provide a new insight into the pathogenesis of AD but also be beneficial for early targeted intervention of AD. This review summarizes the pathogenesis as well as the diagnostic and therapeutic value of genetics in AD.
Collapse
|
125
|
Kuo YC, Rajesh R. A critical overview of therapeutic strategy and advancement for Alzheimer's disease treatment. J Taiwan Inst Chem Eng 2017. [DOI: 10.1016/j.jtice.2017.05.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
126
|
The protection of novel 2-arylethenylquinoline derivatives against impairment of associative learning memory induced by neural Aβ in C. elegans Alzheimer’s disease model. Neurochem Res 2017; 42:3061-3072. [DOI: 10.1007/s11064-017-2339-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022]
|
127
|
Abstract
IMPORTANCE To provide a comprehensive review of knowledge of the genomics of Alzheimer disease (AD) and DNA amyloid β 42 (Aβ42) vaccination as a potential therapy. OBSERVATIONS Genotype-phenotype correlations of AD are presented to provide a comprehensive appreciation of the spectrum of disease causation. Alzheimer disease is caused in part by the overproduction and lack of clearance of Aβ protein. Oligomer Aβ, the most toxic species of Aβ, causes direct injury to neurons, accompanied by enhanced neuroinflammation, astrocytosis and gliosis, and eventually neuronal loss. The strongest genetic evidence supporting this hypothesis derives from mutations in the amyloid precursor protein (APP) gene. A detrimental APP mutation at the β-secretase cleavage site linked to early-onset AD found in a Swedish pedigree enhances Aβ production, in contrast to a beneficial mutation 2 residues away in APP that reduces Aβ production and protects against the onset of sporadic AD. A number of common variants associated with late-onset AD have been identified including apolipoprotein E, BIN1, ABC7, PICALM, MS4A4E/MS4A6A, CD2Ap, CD33, EPHA1, CLU, CR1, and SORL1. One or 2 copies of the apolipoprotein E ε4 allele are a major risk factor for late-onset AD. With DNA Aβ42 vaccination, a Th2-type noninflammatory immune response was achieved with a downregulation of Aβ42-specific effector (Th1, Th17, and Th2) cell responses at later immunization times. DNA Aβ42 vaccination upregulated T regulator cells (CD4+, CD25+, and FoxP3+) and its cytokine interleukin 10, resulting in downregulation of T effectors. CONCLUSIONS AND RELEVANCE Mutations in APP and PS-1 and PS-2 genes that are associated with early-onset, autosomal, dominantly inherited AD, in addition to the at-risk gene polymorphisms responsible for late-onset AD, all indicate a direct and early role of Aβ in the pathogenesis of AD. A translational result of genomic research has been Aβ-reducing therapies including DNA Aβ42 vaccination as a promising approach to delay or prevent this disease.
Collapse
Affiliation(s)
- Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas2Editor, JAMA Neurology
| | | | - Gang Yu
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas
| | - Weiming Xia
- Geriatric Research, Education and Clinical Center, Bedford Veterans Hospital, Bedford, Massachusetts5Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
128
|
Mehdizadeh M, Hashem Dabaghian F, Shojaee A, Molavi N, Taslimi Z, Shabani R, Soleimani Asl S. Protective Effects of Cyperus Rotundus Extract on Amyloid β-Peptide (1-40)-Induced Memory Impairment in Male Rats: A Behavioral Study. Basic Clin Neurosci 2017; 8:249-254. [PMID: 28781732 PMCID: PMC5535330 DOI: 10.18869/nirp.bcn.8.3.249] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION The Alzheimer Disease (AD) is the most common form of dementia that leads to memory impairment. As the oxidative stress plays an important role in AD pathogenesis, the current study aimed at examining the protective effects of Cyperus Rotundus as an antioxidant on amyloid β (Aβ)-induced memory impairment. METHODS Twenty-eight Wistar male rats received intrahippocampal (IHP) injection of the Aβ (1-40) and C. rotundus (400 mg/kg, intraperitoneally). Spatial memory was assessed by the Morris water-maze (MWM) task. RESULTS In the MWM, Aβ (1-40) significantly increased escape latency and traveled distance (P<0.001). The administration of C. rotundus attenuated the Aβ-induced memory impairment in the MWM task. CONCLUSION The current study findings showed that C. Rotundus could improve the learning impairment, following the Aβ treatment, and it may lead to an improvement of AD-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Mehdi Mehdizadeh
- Research Institute for Islamic and Complementary Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fataneh Hashem Dabaghian
- Research Institute for Islamic and Complementary Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Asie Shojaee
- Research Institute for Islamic and Complementary Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nima Molavi
- Department of Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Taslimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamedan, Iran
| | - Ronak Shabani
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Soleimani Asl
- Endometrium and Endometriosis Research Center, Hamedan University of Medical Sciences, Hamedan, Iran
| |
Collapse
|
129
|
Lin HC, Ho MY, Tsen CM, Huang CC, Wu CC, Huang YJ, Hsiao IL, Chuang CY. From the Cover: Comparative Proteomics Reveals Silver Nanoparticles Alter Fatty Acid Metabolism and Amyloid Beta Clearance for Neuronal Apoptosis in a Triple Cell Coculture Model of the Blood–Brain Barrier. Toxicol Sci 2017; 158:151-163. [DOI: 10.1093/toxsci/kfx079] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
130
|
Šimić G, Babić Leko M, Wray S, Harrington CR, Delalle I, Jovanov-Milošević N, Bažadona D, Buée L, de Silva R, Di Giovanni G, Wischik CM, Hof PR. Monoaminergic neuropathology in Alzheimer's disease. Prog Neurobiol 2017; 151:101-138. [PMID: 27084356 PMCID: PMC5061605 DOI: 10.1016/j.pneurobio.2016.04.001] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/09/2016] [Accepted: 04/05/2016] [Indexed: 01/02/2023]
Abstract
None of the proposed mechanisms of Alzheimer's disease (AD) fully explains the distribution patterns of the neuropathological changes at the cellular and regional levels, and their clinical correlates. One aspect of this problem lies in the complex genetic, epigenetic, and environmental landscape of AD: early-onset AD is often familial with autosomal dominant inheritance, while the vast majority of AD cases are late-onset, with the ε4 variant of the gene encoding apolipoprotein E (APOE) known to confer a 5-20 fold increased risk with partial penetrance. Mechanisms by which genetic variants and environmental factors influence the development of AD pathological changes, especially neurofibrillary degeneration, are not yet known. Here we review current knowledge of the involvement of the monoaminergic systems in AD. The changes in the serotonergic, noradrenergic, dopaminergic, histaminergic, and melatonergic systems in AD are briefly described. We also summarize the possibilities for monoamine-based treatment in AD. Besides neuropathologic AD criteria that include the noradrenergic locus coeruleus (LC), special emphasis is given to the serotonergic dorsal raphe nucleus (DRN). Both of these brainstem nuclei are among the first to be affected by tau protein abnormalities in the course of sporadic AD, causing behavioral and cognitive symptoms of variable severity. The possibility that most of the tangle-bearing neurons of the LC and DRN may release amyloid β as well as soluble monomeric or oligomeric tau protein trans-synaptically by their diffuse projections to the cerebral cortex emphasizes their selective vulnerability and warrants further investigations of the monoaminergic systems in AD.
Collapse
Affiliation(s)
- Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Selina Wray
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | | | - Ivana Delalle
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Nataša Jovanov-Milošević
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Danira Bažadona
- Department of Neurology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Luc Buée
- University of Lille, Inserm, CHU-Lille, UMR-S 1172, Alzheimer & Tauopathies, Lille, France
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Claude M Wischik
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Patrick R Hof
- Fishberg Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
131
|
Shi H, Lee JY. Tautomeric Effect of Histidine on the Monomeric Structure of Amyloid β-Peptide(1-42). ACS Chem Neurosci 2017; 8:669-675. [PMID: 28292182 DOI: 10.1021/acschemneuro.6b00375] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Tautomeric state of histidine is one of the factors that influence the structural and aggregation properties of amyloid β (Aβ)-peptide in neutral state. It is worth it to uncover the monomeric properties of Aβ(1-42) peptide in comparison with Aβ(1-40) peptide. Our replica-exchange molecular dynamics simulations results show that the sheet content of each tautomeric isomer in Aβ(1-42) monomer is slightly higher than that in Aβ(1-40) monomer except His6(δ)-His13(δ)-His14(δ) (δδδ) isomer, implying higher aggregation tendency in Aβ(1-42), which is in agreement with previous experimental and theoretical studies. Further analysis indicates that (εεε), (εδε), (εδδ), and (δδε) isomers prefer sheet conformation although they are in nondominating states. Particularly, it is confirmed that antiparallel β-sheets of (εδδ) were formed at K16-E22 (22.0-43.9%), N27-A30 except G29 (21.9-40.2%), and M35-I41 except G37 (24.1-43.4%). Furthermore, (εδδ) may be the easiest one to overcome structural transformation due to nonobstructing interactions between K16 and/or L17 and histidine residues. The current study will help to understand the tautomeric effect of Aβ(1-42) peptide to overcome Alzheimer's disease.
Collapse
Affiliation(s)
- Hu Shi
- Department of Chemistry, Sungkyunkwan University, Suwon 440-746, Korea
| | - Jin Yong Lee
- Department of Chemistry, Sungkyunkwan University, Suwon 440-746, Korea
| |
Collapse
|
132
|
Dutta M, Mattaparthi VSK. In silico investigation on the inhibition of Aβ 42 aggregation by Aβ 40 peptide by potential of mean force study. J Biomol Struct Dyn 2017; 36:741-752. [PMID: 28278027 DOI: 10.1080/07391102.2017.1296783] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Recent experimental data revealed that small, soluble Amyloid beta (Aβ42) oligomers, especially dimers impair synaptic plasticity and memory leading to Alzheimer's disease. Here, we have studied dimerization of Aβ42/Aβ42 homo-dimer and Aβ40/Aβ42 hetero-dimer in terms of free energy profile by all-atom simulations using the ff99SB force field. We have found that in the presence of Aβ40 peptide, there exists a strong tendency to form a hetero-dimer with Aβ42 peptide, suggesting that a possible co-oligomerization. Furthermore, we have investigated the effects of Aβ40 on the Aβ42 peptide. Our study also shows that in presence of Aβ40, the beta-content of Aβ42 monomer is reduced. Additionally, certain residues important for bending in Aβ42 peptide attained an increased flexibility in the presence of Aβ40. The salt-bridge destabilization also manifested the impact of Aβ40 on Aβ42 peptide as a whole. Based on this, one may expect that Aβ40 inhibits the aggregation propensity of Aβ42. Moreover, the binding free energy obtained by the molecular mechanics-Poisson-Boltzmann surface area method also revealed a strong affinity between the two isoforms thereby suggests that Aβ40 binding induces conformational change in Aβ42. Our results suggest that co-oligomerization of Aβ isoforms may play a substantial role in Alzheimer's disease.
Collapse
Affiliation(s)
- Mary Dutta
- a Department of Molecular Biology and Biotechnology , Tezpur University , Tezpur 784 028 , Assam , India
| | | |
Collapse
|
133
|
Lynn SA, Keeling E, Munday R, Gabha G, Griffiths H, Lotery AJ, Ratnayaka JA. The complexities underlying age-related macular degeneration: could amyloid beta play an important role? Neural Regen Res 2017; 12:538-548. [PMID: 28553324 PMCID: PMC5436342 DOI: 10.4103/1673-5374.205083] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Age-related macular degeneration (AMD) causes irreversible loss of central vision for which there is no effective treatment. Incipient pathology is thought to occur in the retina for many years before AMD manifests from midlife onwards to affect a large proportion of the elderly. Although genetic as well as non-genetic/environmental risks are recognized, its complex aetiology makes it difficult to identify susceptibility, or indeed what type of AMD develops or how quickly it progresses in different individuals. Here we summarize the literature describing how the Alzheimer's-linked amyloid beta (Aβ) group of misfolding proteins accumulate in the retina. The discovery of this key driver of Alzheimer's disease in the senescent retina was unexpected and surprising, enabling an altogether different perspective of AMD. We argue that Aβ fundamentally differs from other substances which accumulate in the ageing retina, and discuss our latest findings from a mouse model in which physiological amounts of Aβ were subretinally-injected to recapitulate salient features of early AMD within a short period. Our discoveries as well as those of others suggest the pattern of Aβ accumulation and pathology in donor aged/AMD tissues are closely reproduced in mice, including late-stage AMD phenotypes, which makes them highly attractive to study dynamic aspects of Aβ-mediated retinopathy. Furthermore, we discuss our findings revealing how Aβ behaves at single-cell resolution, and consider the long-term implications for neuroretinal function. We propose Aβ as a key element in switching to a diseased retinal phenotype, which is now being used as a biomarker for late-stage AMD.
Collapse
Affiliation(s)
- Savannah A Lynn
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Eloise Keeling
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Rosie Munday
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Gagandeep Gabha
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Helen Griffiths
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Andrew J Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Eye Unit, University Southampton NHS Trust, Southampton, United Kingdom
| | - J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
134
|
Affiliation(s)
- Rodrigo Aguayo-Ortiz
- Departamento de Fisicoquímica; Universidad Nacional Autónoma de México; Ciudad de México 04510 México
| | - Laura Dominguez
- Departamento de Fisicoquímica; Universidad Nacional Autónoma de México; Ciudad de México 04510 México
| |
Collapse
|
135
|
Janssen L, Dubbelaar ML, Holtman IR, de Boer-Bergsma J, Eggen BJL, Boddeke HWGM, De Deyn PP, Van Dam D. Aging, microglia and cytoskeletal regulation are key factors in the pathological evolution of the APP23 mouse model for Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2016; 1863:395-405. [PMID: 27838490 DOI: 10.1016/j.bbadis.2016.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 10/21/2016] [Accepted: 11/08/2016] [Indexed: 02/01/2023]
Abstract
Aging is the key risk factor for Alzheimer's disease (AD). In addition, the amyloid-beta (Aβ) peptide is considered a critical neurotoxic agent in AD pathology. However, the connection between these factors is unclear. We aimed to provide an extensive characterization of the gene expression profiles of the amyloidosis APP23 model for AD and control mice and to evaluate the effect of aging on these profiles. We also correlated our findings to changes in soluble Aβ-levels and other pathological and symptomatic features of the model. We observed a clear biphasic expression profile. The first phase displayed a maturation profile, which resembled features found in young carriers of familial AD mutations. The second phase reflected aging processes and showed similarities to the progression of human AD pathology. During this phase, the model displayed a clear upregulation of microglial activation and lysosomal pathways and downregulation of neuron differentiation and axon guidance pathways. Interestingly, the changes in expression were all correlated to aging in general, but appeared more extensive/accelerated in APP23 mice.
Collapse
Affiliation(s)
- Leen Janssen
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Marissa L Dubbelaar
- Department of Neuroscience, Medical Physiology Section, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Inge R Holtman
- Department of Neuroscience, Medical Physiology Section, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Jelkje de Boer-Bergsma
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Bart J L Eggen
- Department of Neuroscience, Medical Physiology Section, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Hendrikus W G M Boddeke
- Department of Neuroscience, Medical Physiology Section, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Peter P De Deyn
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium; University of Groningen, University Medical Center Groningen (UMCG), Department of Neurology and Alzheimer Research Center, Groningen, The Netherlands; Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium.
| |
Collapse
|
136
|
Maternal separation exacerbates Alzheimer's disease-like behavioral and pathological changes in adult APPswe/PS1dE9 mice. Behav Brain Res 2016; 318:18-23. [PMID: 27771383 DOI: 10.1016/j.bbr.2016.10.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 10/17/2016] [Accepted: 10/19/2016] [Indexed: 11/20/2022]
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disorder that gradually destroys memory and cognitive abilities in the elderly, makes a huge emotional and economic burden on the patients and their families. The presence of senile plaques and the loss of cholinergic neurons in the brain are two neuropathological hallmarks of AD. Maternal separation (MS) is an animal paradigm designed to make early life stress. Studies on wild type rodents showed that MS could induce AD-like cognitive deficit and pathological changes. However, the effects of MS on AD susceptible population or AD animal models are still unclear. In the present study, male APPswe/PS1dE9 transgenic mice were separated from dam and pups 3h per day from postnatal day 2 to day 21. After weaning, all animals were housed under normal conditions (4 mice per cage). At 9-month age, MWM tests were performed to evaluate the learning and memory abilities. Then the pathological changes in the brain were measured by histology staining. The results showed MS mice had more severe deficit of learning and memory. Compared to the control, there were more senile plaques in cortex and hippocampus, fewer cholinergic neurons in nucleus basalis of Meynert in MS mice. These results indicate that MS exacerbates Alzheimer's disease-like behavioral and pathological changes in APPswe/PS1dE9 mice.
Collapse
|
137
|
Taylor-Walker G, Lynn SA, Keeling E, Munday R, Johnston DA, Page A, Scott JA, Goverdhan S, Lotery AJ, Ratnayaka JA. The Alzheimer's-related amyloid beta peptide is internalised by R28 neuroretinal cells and disrupts the microtubule associated protein 2 (MAP-2). Exp Eye Res 2016; 153:110-121. [PMID: 27751744 PMCID: PMC5131630 DOI: 10.1016/j.exer.2016.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/12/2016] [Accepted: 10/11/2016] [Indexed: 11/15/2022]
Abstract
Age-related Macular Degeneration (AMD) is a common, irreversible blinding condition that leads to the loss of central vision. AMD has a complex aetiology with both genetic as well as environmental risks factors, and share many similarities with Alzheimer's disease. Recent findings have contributed significantly to unravelling its genetic architecture that is yet to be matched by molecular insights. Studies are made more challenging by observations that aged and AMD retinas accumulate the highly pathogenic Alzheimer's-related Amyloid beta (Aβ) group of peptides, for which there appears to be no clear genetic basis. Analyses of human donor and animal eyes have identified retinal Aβ aggregates in retinal ganglion cells (RGC), the inner nuclear layer, photoreceptors as well as the retinal pigment epithelium. Aβ is also a major drusen constituent; found correlated with elevated drusen-load and age, with a propensity to aggregate in retinas of advanced AMD. Despite this evidence, how such a potent driver of neurodegeneration might impair the neuroretina remains incompletely understood, and studies into this important aspect of retinopathy remains limited. In order to address this we exploited R28 rat retinal cells which due to its heterogeneous nature, offers diverse neuroretinal cell-types in which to study the molecular pathology of Aβ. R28 cells are also unaffected by problems associated with the commonly used RGC-5 immortalised cell-line, thus providing a well-established model in which to study dynamic Aβ effects at single-cell resolution. Our findings show that R28 cells express key neuronal markers calbindin, protein kinase C and the microtubule associated protein-2 (MAP-2) by confocal immunofluorescence which has not been shown before, but also calretinin which has not been reported previously. For the first time, we reveal that retinal neurons rapidly internalised Aβ1-42, the most cytotoxic and aggregate-prone amongst the Aβ family. Furthermore, exposure to physiological amounts of Aβ1-42 for 24 h correlated with impairment to neuronal MAP-2, a cytoskeletal protein which regulates microtubule dynamics in axons and dendrites. Disruption to MAP-2 was transient, and had recovered by 48 h, although internalised Aβ persisted as discrete puncta for as long as 72 h. To assess whether Aβ could realistically localise to living retinas to mediate such effects, we subretinally injected nanomolar levels of oligomeric Aβ1-42 into wildtype mice. Confocal microscopy revealed the presence of focal Aβ deposits in RGC, the inner nuclear and the outer plexiform layers 8 days later, recapitulating naturally-occurring patterns of Aβ aggregation in aged retinas. Our novel findings describe how retinal neurons internalise Aβ to transiently impair MAP-2 in a hitherto unreported manner. MAP-2 dysfunction is reported in AMD retinas, and is thought to be involved in remodelling and plasticity of post-mitotic neurons. Our insights suggest a molecular pathway by which this could occur in the senescent eye leading to complex diseases such as AMD. Molecular basis of complex retinopathies such as AMD is incompletely understood. The Alzheimer's-related Aβ peptides are rapidly internalised by retinal neurons. Internalised Aβ is retained within neurons and transiently impairs MAP-2. Subretinally injected Aβ mimics its naturally-occurring distribution in aged retinas.
Collapse
Affiliation(s)
- George Taylor-Walker
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SGH, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Savannah A Lynn
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SGH, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Eloise Keeling
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SGH, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Rosie Munday
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SGH, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - David A Johnston
- Biomedical Imaging Unit, University of Southampton, SGH, MP12, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Anton Page
- Biomedical Imaging Unit, University of Southampton, SGH, MP12, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Jennifer A Scott
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SGH, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Srini Goverdhan
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SGH, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - Andrew J Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SGH, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom; Eye Unit, University Southampton NHS Trust, Southampton, SO16 6YD, United Kingdom
| | - J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SGH, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom.
| |
Collapse
|
138
|
Fernandez MA, Biette KM, Dolios G, Seth D, Wang R, Wolfe MS. Transmembrane Substrate Determinants for γ-Secretase Processing of APP CTFβ. Biochemistry 2016; 55:5675-5688. [PMID: 27649271 DOI: 10.1021/acs.biochem.6b00718] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The amyloid β-peptide (Aβ) of Alzheimer's disease (AD) is generated by proteolysis within the transmembrane domain (TMD) of a C-terminal fragment of the amyloid β protein-precursor (APP CTFβ) by the γ-secretase complex. This processing produces Aβ ranging from 38 to 49 residues in length. Evidence suggests that this spectrum of Aβ peptides is the result of successive γ-secretase cleavages, with endoproteolysis first occurring at the ε sites to generate Aβ48 or Aβ49, followed by C-terminal trimming mostly every three residues along two product lines to generate shorter, secreted forms of Aβ: the primary Aβ49-46-43-40 line and a minor Aβ48-45-42-38 line. The major secreted Aβ species are Aβ40 and Aβ42, and an increased proportion of the longer, aggregation-prone Aβ42 compared to Aβ40 is widely thought to be important in AD pathogenesis. We examined TMD substrate determinants of the specificity and efficiency of ε site endoproteolysis and carboxypeptidase trimming of CTFβ by γ-secretase. We determined that the C-terminal negative charge of the intermediate Aβ49 does not play a role in its trimming by γ-secretase. Peptidomimetic probes suggest that γ-secretase has S1', S2', and S3' pockets, through which trimming by tripeptides may be determined. However, deletion of residues around the ε sites demonstrates that a depth of three residues within the TMD is not a determinant of the location of endoproteolytic ε cleavage of CTFβ. We also show that instability of the CTFβ TMD helix near the ε site significantly increases endoproteolysis, and that helical instability near the carboxypeptidase cleavage sites facilitates C-terminal trimming by γ-secretase. In addition, we found that CTFβ dimers are not endoproteolyzed by γ-secretase. These results support a model in which initial interaction of the array of residues along the undimerized single helical TMD of substrates dictates the site of initial ε cleavage and that helix unwinding is essential for both endoproteolysis and carboxypeptidase trimming.
Collapse
Affiliation(s)
- Marty A Fernandez
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Kelly M Biette
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Georgia Dolios
- Icahn School of Medicine at Mount Sinai , New York, New York 10029, United States
| | - Divya Seth
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Rong Wang
- Icahn School of Medicine at Mount Sinai , New York, New York 10029, United States
| | - Michael S Wolfe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts 02115, United States
| |
Collapse
|
139
|
Li QX, Cappai R, Evin G, Tanner JE, Gray CW, Beyreuther K, Masters CL. Products of the Alzheimer's disease amyloid precursor protein generated by,β-secretase are present in human platelets, and secreted upon degranulation. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/153331759801300504] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Proteolytic processing of Alzheimer's disease amyloid precursor protein (APP) by /3-secretase generates the Nterminus ofA/3 (which deposits in the brain) and releases a secreted ectodomain of APP (sAPP/3). We identified in human platelets a band at 125 kDa corresponding to APP ectodomain ending with C-terminal methionine residue (APP671) as characterized by an antibody specificfor the C-terminal methionine residue of sAPP/3. The same antibody also detected bands at -105 and U125 kDa in human brain homogenates. Platelet sAPP/3 is an isoform containing the Kunitzprotease inhibitor domain (sAPP/3-KPI+) and is released into the medium when platelets are induced to aggregate using agonists such as thrombin, collagen, phorbol 12-myristate 13-acetate, or calcium ionophore A2318 7. The release of sAPPB /from aggregatedplatelets is consistent with a role in regulation of the coagulation cascade and/or in platelet aggregation. These data together with previous reports suggest that human platelets contain the a-, /3-and y-secretase activities, and are a suitable system to study APP processing and Ap production, a pathway which is considered to be a prime targetfor therapeutic intervention in AD.
Collapse
Affiliation(s)
| | | | | | - Jane E. Tanner
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Carol W. Gray
- SmithKline Beecham Pharmaceuticals, Harlow, Essex, England
| | | | - Colin L. Masters
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
140
|
Day GS, Musiek ES, Roe CM, Norton J, Goate AM, Cruchaga C, Cairns NJ, Morris JC. Phenotypic Similarities Between Late-Onset Autosomal Dominant and Sporadic Alzheimer Disease: A Single-Family Case-Control Study. JAMA Neurol 2016; 73:1125-32. [PMID: 27454811 PMCID: PMC5025942 DOI: 10.1001/jamaneurol.2016.1236] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
IMPORTANCE The amyloid hypothesis posits that disrupted β-amyloid homeostasis initiates the pathological process resulting in Alzheimer disease (AD). Autosomal dominant AD (ADAD) has an early symptomatic onset and is caused by single-gene mutations that result in overproduction of β-amyloid 42. To the extent that sporadic late-onset AD (LOAD) also results from dysregulated β-amyloid 42, the clinical phenotypes of ADAD and LOAD should be similar when controlling for the effects of age. OBJECTIVE To use a family with late-onset ADAD caused by a presenilin 1 (PSEN1) gene mutation to mitigate the potential confound of age when comparing ADAD and LOAD. DESIGN, SETTING, AND PARTICIPANTS This case-control study was conducted at the Knight Alzheimer Disease Research Center at Washington University, St Louis, Missouri, and other National Institutes of Aging-funded AD centers in the United States. Ten PSEN1 A79V mutation carriers from multiple generations of a family with late-onset ADAD and 12 noncarrier family members were followed up at the Knight Alzheimer Disease Research Center (1985-2015) and 1115 individuals with neuropathologically confirmed LOAD were included from the National Alzheimer Coordinating Center database (September 2005-December 2014). Data analysis was completed in January 2016, including Knight Alzheimer Disease Research Center patient data collected up until the end of 2015. MAIN OUTCOMES AND MEASURES Planned comparison of clinical characteristics between cohorts, including age at symptom onset, associated symptoms and signs, rates of progression, and disease duration. RESULTS Of the PSEN1 A79V carriers in the family with late-onset ADAD, 4 were female (57%); among those with LOAD, 529 were female (47%). Seven mutation carriers (70%) developed AD dementia, while 3 were yet asymptomatic in their seventh and eighth decades of life. No differences were observed between mutation carriers and individuals with LOAD concerning age at symptom onset (mutation carriers: mean, 75 years [range, 63-77 years] vs those with LOAD: mean, 74 years [range, 60-101 years]; P = .29), presenting symptoms (memory loss in 7 of 7 mutation carriers [100%] vs 958 of 1063 individuals with LOAD [90.1%]; P ≥ .99) and duration (mutation carriers: mean, 9.9 years [range, 2.3-12.8 years] vs those with LOAD: 9 years [range, 1-27 years]; P = .73), and rate of progression of dementia (median annualized change in Clinical Dementia Rating-Sum of Boxes score, mutation carriers: 1.2 [range, 0.1-3.3] vs those with LOAD: 1.9 [range, -3.5 to 11.9]; P = .73). Early emergence of comorbid hallucinations and delusions were observed in 57% of individuals with ADAD (4 of 7) vs 19% of individuals with LOAD (137 of 706) (P = .03). Three of 12 noncarriers (25%) from the PSEN1 A79V family are potential phenocopies as they also developed AD dementia (median age at onset, 76.0 years). CONCLUSIONS AND RELEVANCE In this family, the amyloidogenic PSEN1 A79V mutation recapitulates the clinical attributes of LOAD. Previously reported clinical phenotypic differences between individuals with ADAD and LOAD may reflect age- or mutation-dependent effects.
Collapse
Affiliation(s)
- Gregory S Day
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Erik S Musiek
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Catherine M Roe
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Joanne Norton
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri3Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Alison M Goate
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri3Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Carlos Cruchaga
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri3Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Nigel J Cairns
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri4Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - John C Morris
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
141
|
Sarroca S, Molina-Martínez P, Aresté C, Etzrodt M, García de Frutos P, Gasa R, Antonell A, Molinuevo JL, Sánchez-Valle R, Saura CA, Lladó A, Sanfeliu C. Preservation of cell-survival mechanisms by the presenilin-1 K239N mutation may cause its milder clinical phenotype. Neurobiol Aging 2016; 46:169-79. [PMID: 27498054 DOI: 10.1016/j.neurobiolaging.2016.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 07/04/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022]
Abstract
Presenilin 1 (PSEN1) mutations are the main cause of monogenic Alzheimer's disease. We studied the functional effects of the mutation K239N, which shows incomplete penetrance at the age of 65 years and compared it with the more aggressive mutation E120G. We engineered stable cell lines expressing human PSEN1 wild type or with K239N or E120G mutations. Both mutations induced dysfunction of γ-secretase in the processing of amyloid-β protein precursor, leading to an increase in the amyloid β42/amyloid β40 ratio. Analysis of homeostatic mechanisms showed that K239N induced lower basal and hydrogen peroxide induced intracellular levels of reactive oxygen species than E120G. Similarly, K239N induced lower vulnerability to apoptosis by hydrogen peroxide injury than E120G. Accordingly, the proapoptotic signaling pathways c-Jun NH2-terminal kinase and p38 mitogen-activated protein kinase maintained PSEN1-mediated negative regulation in K239N but not in E120G-bearing cells. Furthermore, the activation of the prosurvival signaling pathways mitogen-activated protein kinase/extracellular signal-regulated kinase and phosphoinositide 3-kinase/Akt was lower in E120G-bearing cells. Therefore, preservation of mechanisms regulating cell responses independent of amyloid-β protein precursor processing may account for the milder phenotype induced by the PSEN1 K239N mutation.
Collapse
Affiliation(s)
- Sara Sarroca
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC, Barcelona, Spain
| | | | - Cristina Aresté
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC, Barcelona, Spain
| | - Martin Etzrodt
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Pablo García de Frutos
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC, Barcelona, Spain; Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rosa Gasa
- Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Anna Antonell
- Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Alzheimer's Disease and Other Cognitive Disorders Unit, Department of Neurology, Hospital Clínic, Barcelona, Spain
| | - José Luís Molinuevo
- Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Alzheimer's Disease and Other Cognitive Disorders Unit, Department of Neurology, Hospital Clínic, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Alzheimer's Disease and Other Cognitive Disorders Unit, Department of Neurology, Hospital Clínic, Barcelona, Spain
| | - Carlos A Saura
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Albert Lladó
- Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Alzheimer's Disease and Other Cognitive Disorders Unit, Department of Neurology, Hospital Clínic, Barcelona, Spain.
| | - Coral Sanfeliu
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC, Barcelona, Spain; Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
142
|
Atwood CS, Bowen RL. A Unified Hypothesis of Early- and Late-Onset Alzheimer's Disease Pathogenesis. J Alzheimers Dis 2016; 47:33-47. [PMID: 26402752 DOI: 10.3233/jad-143210] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Early-onset familial Alzheimer's disease (EOFAD) and late-onset sporadic AD (LOSAD) both follow a similar pathological and biochemical course that includes: neuron and synapse loss and dysfunction, microvascular damage, microgliosis, extracellular amyloid-β deposition, tau phosphorylation, formation of intracellular neurofibrillary tangles, endoreduplication and related cell cycle events in affected brain regions. Any mechanistic explanation of AD must accommodate these biochemical and neuropathological features for both forms of the disease. In this insight paper we provide a unifying hypothesis for EOFAD and LOSAD that proposes that the aberrant re-entry of terminally differentiated, post-mitotic neurons into the cell division cycle is a common pathway that explains both early and late-onset forms of AD. Cell cycle abnormalities appear very early in the disease process, prior to the appearance of plaques and tangles, and explain the biochemical (e.g. tau phosphorylation), neuropathological (e.g. neuron hypertrophy; polypoidy) and cognitive changes observed in EOFAD and LOSAD. Genetic mutations in AβPP, PSEN1, and PSEN2 that alter amyloid-β precursor protein and Notch processing drive reactivation of the cell cycle in EOFAD, while age-related reproductive endocrine dyscrasia that upregulates mitogenic TNF signaling and AβPP processing toward the amyloidogenic pathway drives reactivation of the cell cycle in LOSAD. In essence, AβPP and presenilin mutations initiate early, what endocrine dyscrasia initiates later: aberrant cell cycle re-entry of post-mitotic neurons leading to neurodegeneration and cognitive decline in AD. Inhibition of cell cycle re-entry in post-mitotic neurons may be a useful therapeutic strategy to prevent, slow or halt disease progression.
Collapse
Affiliation(s)
- Craig S Atwood
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research, Education and Clinical Center, Veterans Administration Hospital, Madison, WI, USA.,School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | | |
Collapse
|
143
|
Iljina M, Garcia GA, Dear AJ, Flint J, Narayan P, Michaels TCT, Dobson CM, Frenkel D, Knowles TPJ, Klenerman D. Quantitative analysis of co-oligomer formation by amyloid-beta peptide isoforms. Sci Rep 2016; 6:28658. [PMID: 27346247 PMCID: PMC4921824 DOI: 10.1038/srep28658] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/07/2016] [Indexed: 01/01/2023] Open
Abstract
Multiple isoforms of aggregation-prone proteins are present under physiological conditions and have the propensity to assemble into co-oligomers with different properties from self-oligomers, but this process has not been quantitatively studied to date. We have investigated the amyloid-β (Aβ) peptide, associated with Alzheimer’s disease, and the aggregation of its two major isoforms, Aβ40 and Aβ42, using a statistical mechanical modelling approach in combination with in vitro single-molecule fluorescence measurements. We find that at low concentrations of Aβ, corresponding to its physiological abundance, there is little free energy penalty in forming co-oligomers, suggesting that the formation of both self-oligomers and co-oligomers is possible under these conditions. Our model is used to predict the oligomer concentration and size at physiological concentrations of Aβ and suggests the mechanisms by which the ratio of Aβ42 to Aβ40 can affect cell toxicity. An increased ratio of Aβ42 to Aβ40 raises the fraction of oligomers containing Aβ42, which can increase the hydrophobicity of the oligomers and thus promote deleterious binding to the cell membrane and increase neuronal damage. Our results suggest that co-oligomers are a common form of aggregate when Aβ isoforms are present in solution and may potentially play a significant role in Alzheimer’s disease.
Collapse
Affiliation(s)
- Marija Iljina
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Gonzalo A Garcia
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Alexander J Dear
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Jennie Flint
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Priyanka Narayan
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Thomas C T Michaels
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Daan Frenkel
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Tuomas P J Knowles
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| |
Collapse
|
144
|
Ellerbrock BR, Coscarelli EM, Gurney ME, Geary TG. Screening for Presenilin Inhibitors Using the Free-Living Nematode, Caenorhabditis elegans. ACTA ACUST UNITED AC 2016; 9:147-52. [PMID: 15006138 DOI: 10.1177/1087057103261038] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Caenorhabditis elegans contains 3 homologs of presenilin genes that are associated with Alzheimer s disease. Loss-of-function mutations in C. elegans genes cause a defect in egg laying. In humans, loss of presenilin-1 (PS1) function reduces amyloid-beta peptide processing from the amyloid protein precursor. Worms were screened for compounds that block egg laying, phenocopying presenilin loss of function. To accommodate even relatively high throughput screening, a semi-automated method to quantify egg laying was devised by measuring the chitinase released into the culture medium. Chitinase is released by hatching eggs, but little is shed into the medium from the body cavity of a hermaphrodite with an egg laying deficient ( egl) phenotype. Assay validation involved measuring chitinase release from wild-type C. elegans (N2 strain), sel-12 presenilin loss-of-function mutants, and 2 strains of C. elegans with mutations in the egl-36K+channel gene. Failure to find specific presenilin inhibitors in this collection likely reflects the small number of compounds tested, rather than a flaw in screening strategy. Absent defined biochemical pathways for presenilin, this screening method, which takes advantage of the genetic system available in C. elegans and its historical use for anthelminthic screening, permits an entry into mechanism-based discovery of drugs for Alzheimer s disease. ( Journal of Biomolecular Screening 2004:147-152)
Collapse
|
145
|
Carroll CM, Li YM. Physiological and pathological roles of the γ-secretase complex. Brain Res Bull 2016; 126:199-206. [PMID: 27133790 DOI: 10.1016/j.brainresbull.2016.04.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/19/2016] [Accepted: 04/27/2016] [Indexed: 12/27/2022]
Abstract
Gamma-secretase (GS) is an enzyme complex that cleaves numerous substrates, and it is best known for cleaving amyloid precursor protein (APP) to form amyloid-beta (Aβ) peptides. Aberrant cleavage of APP can lead to Alzheimer's disease, so much research has been done to better understand GS structure and function in hopes of developing therapeutics for Alzheimer's. Therefore, most of the attention in this field has been focused on developing modulators that reduce pathogenic forms of Aβ while leaving Notch and other GS substrates intact, but GS provides multiple avenues of modulation that could improve AD pathology. GS has complex regulation, through its essential subunits and other associated proteins, providing other targets for AD drugs. Therapeutics can also alter GS trafficking and thereby improve cognition, or move beyond Aβ entirely, effecting Notch and neural stem cells. GS also cleaves substrates that affect synaptic morphology and function, presenting another window by which GS modulation could improve AD pathology. Taken together, GS presents a unique cross road for neural processes and an ideal target for AD therapeutics.
Collapse
Affiliation(s)
- Courtney M Carroll
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, NY, United States; Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, NY, United States.
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, NY, United States; Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, NY, United States; Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, NY, United States
| |
Collapse
|
146
|
Gu M, Viles JH. Methionine oxidation reduces lag-times for amyloid-β(1-40) fiber formation but generates highly fragmented fibers. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1260-1269. [PMID: 27108954 DOI: 10.1016/j.bbapap.2016.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/19/2016] [Indexed: 01/06/2023]
Abstract
Oxidative stress and the formation of amyloid plaques containing amyloid-β (Aβ) peptides are two key hallmarks of Alzheimer's disease. A proportion of methionine (Met) at position 35 within Aβ is oxidized to methionine sulphoxide (Met(OX)) within the Alzheimer's plaques. These oxidative processes may be the key to understanding the early stages of Alzheimer's disease. In vitro oxidation of Aβ, by the physiological oxidant H2O2, was monitored using (1)H NMR and mass spectrometry. Here we investigate the effect of Aβ methionine oxidation on fiber formation kinetics and morphology using the amyloid specific fluorescence dye Thioflavin T (ThT) and Transmission Electron Microscopy (TEM). Methionine oxidation reduces the total amount of fibers generated for both dominant forms of Aβ, however there are marked differences in the effect of Met(OX) between Aβ(1-40) and Aβ(1-42). Surprisingly the presence of Met(OX) reduces lag-times for Aβ(1-40) fiber formation but extends lag-times for Aβ(1-42). TEM indicates a change in fiber morphology with a pronounced reduction in fiber length for both methionine oxidized Aβ(1-40) and Aβ(1-42). In contrast, the morphology of preformed amyloid fibers is largely unaffected by the presence of H2O2. Our studies suggest that methionine oxidation promotes highly fragmented fiber assemblies of Aβ. Oxidative stress associated with Alzheimer's disease can cause oxidation of methionine within Aβ and this in turn will influence the complex assembly of Aβ monomer into amyloid fibers, which is likely to impact Aβ toxicity.
Collapse
Affiliation(s)
- Miao Gu
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, United Kingdom
| | - John H Viles
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, United Kingdom
| |
Collapse
|
147
|
Salazar C, Valdivia G, Ardiles ÁO, Ewer J, Palacios AG. Genetic variants associated with neurodegenerative Alzheimer disease in natural models. Biol Res 2016; 49:14. [PMID: 26919851 PMCID: PMC4769573 DOI: 10.1186/s40659-016-0072-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 02/12/2016] [Indexed: 01/05/2023] Open
Abstract
The use of transgenic models for the study of neurodegenerative diseases has made valuable contributions to the field. However, some important limitations, including protein overexpression and general systemic compensation for the missing genes, has caused researchers to seek natural models that show the main biomarkers of neurodegenerative diseases during aging. Here we review some of these models-most of them rodents, focusing especially on the genetic variations in biomarkers for Alzheimer diseases, in order to explain their relationships with variants associated with the occurrence of the disease in humans.
Collapse
Affiliation(s)
- Claudia Salazar
- Facultad de Ciencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.
| | - Gonzalo Valdivia
- Facultad de Ciencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.
| | - Álvaro O Ardiles
- Facultad de Ciencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.
| | - John Ewer
- Facultad de Ciencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.
| | - Adrián G Palacios
- Facultad de Ciencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile. .,Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Pasaje Harrington 287, Playa Ancha, 2360102, Valparaíso, Chile.
| |
Collapse
|
148
|
Šimić G, Babić Leko M, Wray S, Harrington C, Delalle I, Jovanov-Milošević N, Bažadona D, Buée L, de Silva R, Di Giovanni G, Wischik C, Hof PR. Tau Protein Hyperphosphorylation and Aggregation in Alzheimer's Disease and Other Tauopathies, and Possible Neuroprotective Strategies. Biomolecules 2016; 6:6. [PMID: 26751493 PMCID: PMC4808800 DOI: 10.3390/biom6010006] [Citation(s) in RCA: 461] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 11/28/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022] Open
Abstract
Abnormal deposition of misprocessed and aggregated proteins is a common final pathway of most neurodegenerative diseases, including Alzheimer's disease (AD). AD is characterized by the extraneuronal deposition of the amyloid β (Aβ) protein in the form of plaques and the intraneuronal aggregation of the microtubule-associated protein tau in the form of filaments. Based on the biochemically diverse range of pathological tau proteins, a number of approaches have been proposed to develop new potential therapeutics. Here we discuss some of the most promising ones: inhibition of tau phosphorylation, proteolysis and aggregation, promotion of intra- and extracellular tau clearance, and stabilization of microtubules. We also emphasize the need to achieve a full understanding of the biological roles and post-translational modifications of normal tau, as well as the molecular events responsible for selective neuronal vulnerability to tau pathology and its propagation. It is concluded that answering key questions on the relationship between Aβ and tau pathology should lead to a better understanding of the nature of secondary tauopathies, especially AD, and open new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia.
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia.
| | - Selina Wray
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK.
| | - Charles Harrington
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| | - Ivana Delalle
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston 02118, MA, USA.
| | - Nataša Jovanov-Milošević
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia.
| | - Danira Bažadona
- Department of Neurology, University Hospital Center Zagreb, Zagreb 10000, Croatia.
| | - Luc Buée
- Laboratory Alzheimer & Tauopathies, Université Lille and INSERM U1172, Jean-Pierre Aubert Research Centre, Lille 59045, France.
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK.
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta.
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK.
| | - Claude Wischik
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| | - Patrick R Hof
- Fishberg Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
149
|
Josić D, Andjelković U. The Role of Proteomics in Personalized Medicine. Per Med 2016. [DOI: 10.1007/978-3-319-39349-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
150
|
Cardoso BR, Busse AL, Hare DJ, Cominetti C, Horst MA, McColl G, Magaldi RM, Jacob-Filho W, Cozzolino SMF. Pro198Leu polymorphism affects the selenium status and GPx activity in response to Brazil nut intake. Food Funct 2016; 7:825-33. [DOI: 10.1039/c5fo01270h] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Selenoproteins play important roles in antioxidant mechanisms, but it is hypothesised that single polymorphism nucleotides (SNPs) may affect their function.
Collapse
Affiliation(s)
- Bárbara R. Cardoso
- Faculty of Pharmaceutical Sciences
- Department of Food and Experimental Nutrition
- University of São Paulo
- São Paulo
- Brazil
| | - Alexandre L. Busse
- Geriatrics Division
- Department of Internal Medicine
- University of São Paulo Medical School
- São Paulo
- Brazil
| | - Dominic J. Hare
- The Florey Institute of Neuroscience and Mental Health
- The University of Melbourne
- Parkville
- Australia
- Elemental Bio-imaging Facility
| | | | - Maria A. Horst
- Faculty of Pharmaceutical Sciences
- Department of Food and Experimental Nutrition
- University of São Paulo
- São Paulo
- Brazil
| | - Gawain McColl
- The Florey Institute of Neuroscience and Mental Health
- The University of Melbourne
- Parkville
- Australia
| | - Regina M. Magaldi
- Geriatrics Division
- Department of Internal Medicine
- University of São Paulo Medical School
- São Paulo
- Brazil
| | - Wilson Jacob-Filho
- Geriatrics Division
- Department of Internal Medicine
- University of São Paulo Medical School
- São Paulo
- Brazil
| | - Silvia M. F. Cozzolino
- Faculty of Pharmaceutical Sciences
- Department of Food and Experimental Nutrition
- University of São Paulo
- São Paulo
- Brazil
| |
Collapse
|