1
|
Cade S, Prestidge C, Zhou X, Bobrovskaya L. The effects of a bioavailable curcumin formulation on Alzheimer's disease pathologies: A potential risk for neuroinflammation. IBRAIN 2024; 10:500-518. [PMID: 39691427 PMCID: PMC11649387 DOI: 10.1002/ibra.12187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/19/2024]
Abstract
Alzheimer's disease (AD) is a common cause of dementia characterized by the presence of two proteinaceous deposits in the brain. These pathologies may be a consequence of complex interactions between neurons and glia before the onset of cognitive impairments. Curcumin, a bioactive compound found in turmeric, is a promising candidate for AD because it alleviates neuropathologies in mouse models of the disease. Although its clinical efficacy has been hindered by low oral bioavailability, the development of new formulations may overcome this limitation. The purpose of this study was to determine the effects of a bioavailable curcumin formulation in a mouse model of AD. The formulation was administered to mice in drinking water after encapsulation into micelles using a previously validated method. A neuropathological assessment was performed to determine if it slows or alters the course of the disease. Cognitive performance was not included because it had already been assessed by a previous study. The bioavailable curcumin formulation was unable to alter the size or number of amyloid plaques in a transgenic mouse model. In addition, mechanisms that regulate amyloid beta production were unchanged, suggesting that the disease had not been altered. The number of reactive astrocytes in the hippocampus and dentate gyrus was not altered by curcumin. However, protein levels of glial fibrillary acidic protein were increased overall in the brain, suggesting that it may have aggravated neuroinflammation. Therefore, a higher dosage, despite its enhanced oral bioavailability, may have a potential risk for neuroinflammation.
Collapse
Affiliation(s)
- Shaun Cade
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Clive Prestidge
- Center for Pharmaceutical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Xin‐Fu Zhou
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
2
|
Ichimata S, Hata Y, Yoshida K, Hirono K, Nishida N. Distinctive Deposition Patterns of Sporadic Transthyretin-Derived Amyloidosis in the Atria: A Forensic Autopsy-Based Study. Int J Mol Sci 2024; 25:8176. [PMID: 39125745 PMCID: PMC11311325 DOI: 10.3390/ijms25158176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Left-to-right differences in the histopathologic patterns of transthyretin-derived amyloid (ATTR) deposition in the atria of older adults have not yet been investigated. Hence, this study evaluated heart specimens from 325 serial autopsy subjects. The amount of ATTR deposits in the seven cardiac regions, including both sides of atria and atrial appendages, was evaluated semiquantitatively. Using digital pathology, we quantitatively evaluated the immunohistochemical deposition burden of ATTR in the myocardium. We identified 20 sporadic ATTR cardiac amyloidosis cases (nine males). All patients had ATTR deposition in the left atrial regions of the myocardium. In the semiquantitative analysis, 14 of the 20 cases showed more severe ATTR deposition on the left atrial regions than on the right side, with statistically significant differences in the pathology grading (p < 0.01 for both the atrium and atrial appendage). Quantitative analysis further supported the difference. Moreover, six had ATTR deposition in the epineurium and/or neural fibers of the atria. Cluster analysis revealed that ATTR deposition in the myocardium was significantly more severe in males than in females. The heterogeneous distribution of amyloid deposits between atria revealed in this study may impair the orderly transmission of the cardiac conduction system and induce arrhythmias, which may be further aggravated by additional neuropathy in the advanced phase. This impairment could be more severe among males. These findings emphasize that atrial evaluation is important for individuals with sporadic ATTR cardiac amyloidosis, particularly for early detection.
Collapse
Affiliation(s)
- Shojiro Ichimata
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan (N.N.)
| | - Yukiko Hata
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan (N.N.)
| | - Koji Yoshida
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan (N.N.)
| | - Keiichi Hirono
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan;
| | - Naoki Nishida
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan (N.N.)
| |
Collapse
|
3
|
Al Khashali H, Ray R, Darweesh B, Wozniak C, Haddad B, Goel S, Seidu I, Khalil J, Lopo B, Murshed N, Guthrie J, Heyl D, Evans HG. Amyloid Beta Leads to Decreased Acetylcholine Levels and Non-Small Cell Lung Cancer Cell Survival via a Mechanism That Involves p38 Mitogen-Activated Protein Kinase and Protein Kinase C in a p53-Dependent and -Independent Manner. Int J Mol Sci 2024; 25:5033. [PMID: 38732252 PMCID: PMC11084752 DOI: 10.3390/ijms25095033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024] Open
Abstract
Several studies have shown an inverse correlation between the likelihood of developing a neurodegenerative disorder and cancer. We previously reported that the levels of amyloid beta (Aβ), at the center of Alzheimer's disease pathophysiology, are regulated by acetylcholinesterase (AChE) in non-small cell lung cancer (NSCLC). Here, we examined the effect of Aβ or its fragments on the levels of ACh in A549 (p53 wild-type) and H1299 (p53-null) NSCLC cell media. ACh levels were reduced by cell treatment with Aβ 1-42, Aβ 1-40, Aβ 1-28, and Aβ 25-35. AChE and p53 activities increased upon A549 cell treatment with Aβ, while knockdown of p53 in A549 cells increased ACh levels, decreased AChE activity, and diminished the Aβ effects. Aβ increased the ratio of phospho/total p38 MAPK and decreased the activity of PKC. Inhibiting p38 MAPK reduced the activity of p53 in A549 cells and increased ACh levels in the media of both cell lines, while opposite effects were found upon inhibiting PKC. ACh decreased the activity of p53 in A549 cells, decreased p38 MAPK activity, increased PKC activity, and diminished the effect of Aβ on those activities. Moreover, the negative effect of Aβ on cell viability was diminished by cell co-treatment with ACh.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Hedeel Guy Evans
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI 48197, USA; (H.A.K.); (R.R.); (B.D.); (C.W.); (B.H.); (S.G.); (I.S.); (J.K.); (B.L.); (N.M.); (J.G.); (D.H.)
| |
Collapse
|
4
|
Jagadeesan N, Roules GC, Chandrashekar DV, Yang J, Kolluru S, Sumbria RK. Modulation of hippocampal protein expression by a brain penetrant biologic TNF-α inhibitor in the 3xTg Alzheimer's disease mice. J Transl Med 2024; 22:291. [PMID: 38500108 PMCID: PMC10946165 DOI: 10.1186/s12967-024-05008-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Biologic TNF-α inhibitors (bTNFIs) can block cerebral TNF-α in Alzheimer's disease (AD) if these macromolecules can cross the blood-brain barrier (BBB). Thus, a model bTNFI, the extracellular domain of type II TNF-α receptor (TNFR), which can bind to and sequester TNF-α, was fused with a mouse transferrin receptor antibody (TfRMAb) to enable brain delivery via BBB TfR-mediated transcytosis. Previously, we found TfRMAb-TNFR to be protective in a mouse model of amyloidosis (APP/PS1) and tauopathy (PS19), and herein we investigated its effects in mice that combine both amyloidosis and tauopathy (3xTg-AD). METHODS Eight-month-old female 3xTg-AD mice were injected intraperitoneally with saline (n = 11) or TfRMAb-TNFR (3 mg/kg; n = 11) three days per week for 12 weeks. Age-matched wild-type (WT) mice (n = 9) were treated similarly with saline. Brains were processed for immunostaining and high-resolution multiplex NanoString GeoMx spatial proteomics. RESULTS We observed regional differences in proteins relevant to Aβ, tau, and neuroinflammation in the hippocampus of 3xTg-AD mice compared with WT mice. From 64 target proteins studied using spatial proteomics, a comparison of the Aβ-plaque bearing vs. plaque-free regions in the 3xTg-AD mice yielded 39 differentially expressed proteins (DEP) largely related to neuroinflammation (39% of DEP) and Aβ and tau pathology combined (31% of DEP). Hippocampal spatial proteomics revealed that the majority of the proteins modulated by TfRMAb-TNFR in the 3xTg-AD mice were relevant to microglial function (⁓ 33%). TfRMAb-TNFR significantly reduced mature Aβ plaques and increased Aβ-associated microglia around larger Aβ deposits in the 3xTg-AD mice. Further, TfRMAb-TNFR increased mature Aβ plaque-associated microglial TREM2 in 3xTg-AD mice. CONCLUSION Overall, despite the low visual Aβ load in the 11-month-old female 3xTg-AD mice, our results highlight region-specific AD-relevant DEP in the hippocampus of these mice. Chronic TfRMAb-TNFR dosing modulated several DEP involved in AD pathology and showed a largely microglia-centric mechanism of action in the 3xTg-AD mice.
Collapse
Affiliation(s)
- Nataraj Jagadeesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - G Chuli Roules
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Devaraj V Chandrashekar
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Joshua Yang
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA
| | - Sanjana Kolluru
- Rancho Cucamonga High School, 11801 Lark Dr, Rancho Cucamonga, CA, 91701, USA
| | - Rachita K Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, 92618, USA.
- Department of Neurology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
5
|
Haddad B, Khalil J, Al Khashali H, Ray R, Goel S, Darweesh B, Coleman KL, Wozniak C, Ranzenberger R, Lopo B, Guthrie J, Heyl D, Evans HG. The role of leptin in regulation of the soluble amyloid precursor protein α (sAPPα) levels in lung cancer cell media. Sci Rep 2024; 14:4921. [PMID: 38418632 PMCID: PMC10901813 DOI: 10.1038/s41598-024-55717-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024] Open
Abstract
Previously, we found that the levels of soluble amyloid precursor protein α (sAPPα) are regulated, in part, by acetylcholinesterase (AChE) in human A549 (p53 wild-type) and H1299 (p53-null) NSCLC cell lines. In this study, we found regulation of sAPPα levels in the media by leptin, a widely recognized obesity-associated adipokine that has recently been shown to play a possible role in cancer signaling. Increased levels of sAPPα, that were accompanied by lower Aβ40/42 levels in the media of A549 and H1299 cells, were detected upon cell incubation with leptin. Conversely, knockdown of leptin or its receptor led to reduced levels of sAPPα and increased levels of Aβ40/42 in the media of A549 and H1299 cells, suggesting that leptin likely shifts APP processing toward the non-amyloidogenic pathway. A549 cell treatment with leptin increased acetylcholine levels and blocked the activities of AChE and p53. Treatment with leptin resulted in increased activation of PKC, ERK1/2, PI3K, and the levels of sAPPα, effects that were reversed by treatment with kinase inhibitors and/or upon addition of AChE to A549 and H1299 cell media. Cell viability increased by treatment of A549 and H1299 cells with leptin and decreased upon co-treatment with AChE and/or inhibitors targeting PKC, ERK1/2, and PI3K. This study is significant as it provides evidence for a likely carcinogenic role of leptin in NSCLC cells via upregulation of sAPPα levels in the media, and highlights the importance of targeting leptin as a potential therapeutic strategy for NSCLC treatment.
Collapse
Affiliation(s)
- Ben Haddad
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Jeneen Khalil
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Hind Al Khashali
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Ravel Ray
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Stuti Goel
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Ban Darweesh
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Kai-Ling Coleman
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Caroline Wozniak
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Robert Ranzenberger
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Brooke Lopo
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Jeffrey Guthrie
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Deborah Heyl
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Hedeel Guy Evans
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA.
| |
Collapse
|
6
|
Area-Gomez E, Schon EA. Towards a Unitary Hypothesis of Alzheimer's Disease Pathogenesis. J Alzheimers Dis 2024; 98:1243-1275. [PMID: 38578892 DOI: 10.3233/jad-231318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
The "amyloid cascade" hypothesis of Alzheimer's disease (AD) pathogenesis invokes the accumulation in the brain of plaques (containing the amyloid-β protein precursor [AβPP] cleavage product amyloid-β [Aβ]) and tangles (containing hyperphosphorylated tau) as drivers of pathogenesis. However, the poor track record of clinical trials based on this hypothesis suggests that the accumulation of these peptides is not the only cause of AD. Here, an alternative hypothesis is proposed in which the AβPP cleavage product C99, not Aβ, is the main culprit, via its role as a regulator of cholesterol metabolism. C99, which is a cholesterol sensor, promotes the formation of mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a cholesterol-rich lipid raft-like subdomain of the ER that communicates, both physically and biochemically, with mitochondria. We propose that in early-onset AD (EOAD), MAM-localized C99 is elevated above normal levels, resulting in increased transport of cholesterol from the plasma membrane to membranes of intracellular organelles, such as ER/endosomes, thereby upregulating MAM function and driving pathology. By the same token, late-onset AD (LOAD) is triggered by any genetic variant that increases the accumulation of intracellular cholesterol that, in turn, boosts the levels of C99 and again upregulates MAM function. Thus, the functional cause of AD is upregulated MAM function that, in turn, causes the hallmark disease phenotypes, including the plaques and tangles. Accordingly, the MAM hypothesis invokes two key interrelated elements, C99 and cholesterol, that converge at the MAM to drive AD pathogenesis. From this perspective, AD is, at bottom, a lipid disorder.
Collapse
Affiliation(s)
- Estela Area-Gomez
- Department of Neurology, Columbia University, New York, NY, USA
- Centro de Investigaciones Biológicas "Margarita Salas", Spanish National Research Council, Madrid, Spain
| | - Eric A Schon
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Genetics and Development>, Columbia University, New York, NY, USA
| |
Collapse
|
7
|
Ichimata S, Yoshida K, Li J, Rogaeva E, Lang AE, Kovacs GG. The molecular spectrum of amyloid-beta (Aβ) in neurodegenerative diseases beyond Alzheimer's disease. Brain Pathol 2024; 34:e13210. [PMID: 37652560 PMCID: PMC10711260 DOI: 10.1111/bpa.13210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023] Open
Abstract
This study investigated the molecular spectrum of amyloid-beta (Aβ) in neurodegenerative diseases beyond Alzheimer's disease (AD). We analyzed Aβ deposition in the temporal cortex and striatum in 116 autopsies, including Lewy body disease (LBD; N = 51), multiple system atrophy (MSA; N = 10), frontotemporal lobar degeneration-TDP-43 (FTLD-TDP; N = 16), and progressive supranuclear palsy (PSP; N = 39). The LBD group exhibited the most Aβ deposition in the temporal cortex and striatum (90/76%, respectively), followed by PSP (69/28%), FTLD-TDP (50/25%), and the MSA group (50/10%). We conducted immunohistochemical analysis using antibodies targeting eight Aβ epitopes in the LBD and PSP groups. Immunohistochemical findings were evaluated semi-quantitatively and quantitatively using digital pathology. Females with LBD exhibited significantly more severe Aβ deposition, particularly Aβ42 and Aβ43 , along with significantly more severe tau pathology. Furthermore, a quantitative analysis of all Aβ peptides in the LBD group revealed an association with the APOE-ε4 genotypes. No significant differences were observed between males and females in the PSP group. Finally, we compared striatal Aβ deposition in cases with LBD (N = 15), AD without α-synuclein pathology (N = 6), and PSP (N = 5). There were no differences in the pan-Aβ antibody (6F/3D)-immunolabeled deposition burden among the three groups, but the deposition burden of peptides with high aggregation capacity, especially Aβ43 , was significantly higher in the AD and LBD groups than in the PSP group. Furthermore, considerable heterogeneity was observed in the composition of Aβ peptides on a case-by-case basis in the AD and LBD groups, whereas it was relatively uniform in the PSP group. Cluster analysis further supported these findings. Our data suggest that the type of concomitant proteinopathies influences the spectrum of Aβ deposition, impacted also by sex and APOE genotypes.
Collapse
Affiliation(s)
- Shojiro Ichimata
- Tanz Centre for Research in Neurodegenerative DiseaseUniversity of TorontoTorontoOntarioCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
- Department of Legal Medicine, Faculty of MedicineUniversity of ToyamaToyamaJapan
| | - Koji Yoshida
- Tanz Centre for Research in Neurodegenerative DiseaseUniversity of TorontoTorontoOntarioCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
- Department of Legal Medicine, Faculty of MedicineUniversity of ToyamaToyamaJapan
| | - Jun Li
- Tanz Centre for Research in Neurodegenerative DiseaseUniversity of TorontoTorontoOntarioCanada
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative DiseaseUniversity of TorontoTorontoOntarioCanada
| | - Anthony E. Lang
- Tanz Centre for Research in Neurodegenerative DiseaseUniversity of TorontoTorontoOntarioCanada
- Edmond J Safra Program in Parkinson's Disease and Rossy Program in Progressive Supranuclear PalsyToronto Western HospitalTorontoOntarioCanada
| | - Gabor G. Kovacs
- Tanz Centre for Research in Neurodegenerative DiseaseUniversity of TorontoTorontoOntarioCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
- Edmond J Safra Program in Parkinson's Disease and Rossy Program in Progressive Supranuclear PalsyToronto Western HospitalTorontoOntarioCanada
- Laboratory Medicine Program and Krembil Brain InstituteUniversity Health NetworkTorontoOntarioCanada
| |
Collapse
|
8
|
Kwon SH, Parthiban S, Tippani M, Divecha HR, Eagles NJ, Lobana JS, Williams SR, Mak M, Bharadwaj RA, Kleinman JE, Hyde TM, Page SC, Hicks SC, Martinowich K, Maynard KR, Collado-Torres L. Influence of Alzheimer's disease related neuropathology on local microenvironment gene expression in the human inferior temporal cortex. GEN BIOTECHNOLOGY 2023; 2:399-417. [PMID: 39329069 PMCID: PMC11426291 DOI: 10.1089/genbio.2023.0019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Neuropathological lesions in the brains of individuals affected with neurodegenerative disorders are hypothesized to trigger molecular and cellular processes that disturb homeostasis of local microenvironments. Here, we applied the 10x Genomics Visium Spatial Proteogenomics (Visium-SPG) platform, which couples spatial gene expression with immunofluorescence protein co-detection, to evaluate its ability to quantify changes in spatial gene expression with respect to amyloid-β (Aβ) and hyperphosphorylated tau (pTau) pathology in post-mortem human brain tissue from individuals with Alzheimer's disease (AD). We identified transcriptomic signatures associated with proximity to Aβ in the human inferior temporal cortex (ITC) during late-stage AD, which we further investigated at cellular resolution with combined immunofluorescence and single molecule fluorescent in situ hybridization (smFISH). The study provides a data analysis workflow for Visium-SPG, and the data represent a proof-of-principal for the power of multi-omic profiling in identifying changes in molecular dynamics that are spatially-associated with pathology in the human brain. We provide the scientific community with web-based, interactive resources to access the datasets of the spatially resolved AD-related transcriptomes at https://research.libd.org/Visium_SPG_AD/.
Collapse
Affiliation(s)
- Sang Ho Kwon
- The Biochemistry, Cellular, and Molecular Biology Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sowmya Parthiban
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Heena R. Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Nicholas J. Eagles
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Jashandeep S. Lobana
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | | | | | - Rahul A. Bharadwaj
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephanie C. Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Kristen R. Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
9
|
Johansson B, Oasa S, Muntsant Soria A, Tiiman A, Söderberg L, Amandius E, Möller C, Lannfelt L, Terenius L, Giménez-Llort L, Vukojević V. The interwoven fibril-like structure of amyloid-beta plaques in mouse brain tissue visualized using super-resolution STED microscopy. Cell Biosci 2023; 13:142. [PMID: 37542303 PMCID: PMC10403925 DOI: 10.1186/s13578-023-01086-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/14/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Standard neuropathologic analysis of Alzheimer's brain relies on traditional fluorescence microscopy, which suffers from limited spatial resolution due to light diffraction. As a result, it fails to reveal intricate details of amyloid plaques. While electron microscopy (EM) offers higher resolution, its extensive sample preparation, involving fixation, dehydration, embedding, and sectioning, can introduce artifacts and distortions in the complex brain tissue. Moreover, EM lacks molecular specificity and has limited field of view and imaging depth. RESULTS In our study, we employed super-resolution Stimulated Emission Depletion (STED) microscopy in conjunction with the anti-human APP recombinant antibody 1C3 fluorescently labelled with DyLightTM633 (1C3-DyLight633). This combination allowed us to visualize amyloidogenic aggregates in vitro and in brain sections from a 17-month-old 3×Tg-AD mouse with sub-diffraction limited spatial resolution. Remarkably, we achieved a spatial resolution of 29 nm in vitro and 62 nm in brain tissue sections, surpassing the capabilities of conventional confocal microscopy by 5-10 times. Consequently, we could discern individual fibrils within plaques, an achievement previously only possible with EM. CONCLUSIONS The utilization of STED microscopy represents a groundbreaking advancement in the field, enabling researchers to delve into the characterization of local mechanisms that underlie Amyloid (Aβ) deposition into plaques and their subsequent clearance. This unprecedented level of detail is especially crucial for comprehending the etiology of Alzheimer's disease and developing the next generation of anti-amyloid treatments. By facilitating the evaluation of drug candidates and non-pharmacological interventions aiming to reduce amyloid burden, STED microscopy emerges as an indispensable tool for driving scientific progress in Alzheimer's research.
Collapse
Affiliation(s)
- Björn Johansson
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - Sho Oasa
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - Aida Muntsant Soria
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Ann Tiiman
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | | | | | | | | | - Lars Terenius
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - Lydia Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Vladana Vukojević
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden.
| |
Collapse
|
10
|
Ichimata S, Martinez-Valbuena I, Lee S, Li J, Karakani AM, Kovacs GG. Distinct Molecular Signatures of Amyloid-Beta and Tau in Alzheimer's Disease Associated with Down Syndrome. Int J Mol Sci 2023; 24:11596. [PMID: 37511361 PMCID: PMC10380583 DOI: 10.3390/ijms241411596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Limited comparative data exist on the molecular spectrum of amyloid-beta (Aβ) and tau deposition in individuals with Down syndrome (DS) and sporadic Alzheimer's disease (sAD). We assessed Aβ and tau deposition severity in the temporal lobe and cerebellum of ten DS and ten sAD cases. Immunohistochemistry was performed using antibodies against eight different Aβ epitopes (6F/3D, Aβ38, Aβ39, Aβ40, Aβ42, Aβ43, pyroglutamate Aβ at third glutamic acid (AβNp3E), phosphorylated- (p-)Aβ at 8th serine (AβpSer8)), and six different pathological tau epitopes (p-Ser202/Thr205, p-Thr231, p-Ser396, Alz50, MC1, GT38). Findings were evaluated semi-quantitatively and quantitatively using digital pathology. DS cases had significantly higher neocortical parenchymal deposition (Aβ38, Aβ42, and AβpSer8), and cerebellar parenchymal deposition (Aβ40, Aβ42, AβNp3E, and AβpSer8) than sAD cases. Furthermore, DS cases had a significantly larger mean plaque size (6F/3D, Aβ42, AβNp3E) in the temporal lobe, and significantly greater deposition of cerebral and cerebellar Aβ42 than sAD cases in the quantitative analysis. Western blotting corroborated these findings. Regarding tau pathology, DS cases had significantly more severe cerebral tau deposition than sAD cases, especially in the white matter (p-Ser202/Thr205, p-Thr231, Alz50, and MC1). Greater total tau deposition in the white matter (p-Ser202/Thr205, p-Thr231, and Alz50) of DS cases was confirmed by quantitative analysis. Our data suggest that the Aβ and tau molecular signatures in DS are distinct from those in sAD.
Collapse
Affiliation(s)
- Shojiro Ichimata
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama 930-8555, Japan
| | - Ivan Martinez-Valbuena
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
| | - Seojin Lee
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
| | - Jun Li
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
| | - Ali M. Karakani
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
| | - Gabor G. Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Edmond J. Safra Program in Parkinson’s Disease, Rossy Program for PSP Research and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON M5T 2S8, Canada
- Laboratory Medicine Program, Krembil Brain Institute, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
11
|
Villarreal J, Kow K, Pham B, Egatz-Gomez A, Sandrin TR, Coleman PD, Ros A. Intracellular Amyloid-β Detection from Human Brain Sections Using a Microfluidic Immunoassay in Tandem with MALDI-MS. Anal Chem 2023; 95:5522-5531. [PMID: 36894164 PMCID: PMC10078609 DOI: 10.1021/acs.analchem.2c03825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023]
Abstract
Alzheimer's disease (AD) currently affects more than 30 million people worldwide. The lack of understanding of AD's physiopathology limits the development of therapeutic and diagnostic tools. Soluble amyloid-β peptide (Aβ) oligomers that appear as intermediates along the Aβ aggregation into plaques are considered among the main AD neurotoxic species. Although a wealth of data are available about Aβ from in vitro and animal models, there is little known about intracellular Aβ in human brain cells, mainly due to the lack of technology to assess the intracellular protein content. The elucidation of the Aβ species in specific brain cell subpopulations can provide insight into the role of Aβ in AD and the neurotoxic mechanism involved. Here, we report a microfluidic immunoassay for in situ mass spectrometry analysis of intracellular Aβ species from archived human brain tissue. This approach comprises the selective laser dissection of individual pyramidal cell bodies from tissues, their transfer to the microfluidic platform for sample processing on-chip, and mass spectrometric characterization. As a proof-of-principle, we demonstrate the detection of intracellular Aβ species from as few as 20 human brain cells.
Collapse
Affiliation(s)
- Jorvani
Cruz Villarreal
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Center
for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Keegan Kow
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Center
for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Brian Pham
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Center
for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Ana Egatz-Gomez
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Center
for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Todd R. Sandrin
- School
of Mathematical and Natural Sciences, Arizona
State University, Glendale, Arizona 85306, United States
- Julie
Ann Wrigley Global Futures Laboratory, Arizona
State University, Glendale, Arizona 85306, United States
| | - Paul D. Coleman
- Banner
ASU Neurodegenerative Research Center, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Alexandra Ros
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Center
for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
12
|
Sheikh AM, Yano S, Tabassum S, Mitaki S, Michikawa M, Nagai A. Alzheimer's Amyloid β Peptide Induces Angiogenesis in an Alzheimer's Disease Model Mouse through Placental Growth Factor and Angiopoietin 2 Expressions. Int J Mol Sci 2023; 24:ijms24054510. [PMID: 36901941 PMCID: PMC10003449 DOI: 10.3390/ijms24054510] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Increased angiogenesis, especially the pathological type, has been documented in Alzheimer's disease (AD) brains, and it is considered to be activated due to a vascular dysfunction-mediated hypoxic condition. To understand the role of the amyloid β (Aβ) peptide in angiogenesis, we analyzed its effects on the brains of young APP transgenic AD model mice. Immunostaining results revealed that Aβ was mainly localized intracellularly, with very few immunopositive vessels, and there was no extracellular deposition at this age. Solanum tuberosum lectin staining demonstrated that compared to their wild-type littermates, the vessel number was only increased in the cortex of J20 mice. CD105 staining also showed an increased number of new vessels in the cortex, some of which were partially positive for collagen4. Real-time PCR results demonstrated that placental growth factor (PlGF) and angiopoietin 2 (AngII) mRNA were increased in both the cortex and hippocampus of J20 mice compared to their wild-type littermates. However, vascular endothelial growth factor (VEGF) mRNA did not change. Immunofluorescence staining confirmed the increased expression of PlGF and AngII in the cortex of the J20 mice. Neuronal cells were positive for PlGF and AngII. Treatment of a neural stem cell line (NMW7) with synthetic Aβ1-42 directly increased the expression of PlGF and AngII, at mRNA levels, and AngII at protein levels. Thus, these pilot data indicate that pathological angiogenesis exists in AD brains due to the direct effects of early Aβ accumulation, suggesting that the Aβ peptide regulates angiogenesis through PlGF and AngII expression.
Collapse
Affiliation(s)
- Abdullah Md. Sheikh
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
- Correspondence: (A.M.S.); (A.N.); Tel.: +81-0853-20-2306 (A.M.S.); +81-0853-20-2198 (A.N.)
| | - Shozo Yano
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Shatera Tabassum
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Shingo Mitaki
- Department of Neurology, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Makoto Michikawa
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Atsushi Nagai
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
- Department of Neurology, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
- Correspondence: (A.M.S.); (A.N.); Tel.: +81-0853-20-2306 (A.M.S.); +81-0853-20-2198 (A.N.)
| |
Collapse
|
13
|
Busch L, Eggert S, Endres K, Bufe B. The Hidden Role of Non-Canonical Amyloid β Isoforms in Alzheimer's Disease. Cells 2022; 11:3421. [PMID: 36359817 PMCID: PMC9654995 DOI: 10.3390/cells11213421] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 09/08/2024] Open
Abstract
Recent advances have placed the pro-inflammatory activity of amyloid β (Aβ) on microglia cells as the focus of research on Alzheimer's Disease (AD). Researchers are confronted with an astonishing spectrum of over 100 different Aβ variants with variable length and chemical modifications. With the exception of Aβ1-42 and Aβ1-40, the biological significance of most peptides for AD is as yet insufficiently understood. We therefore aim to provide a comprehensive overview of the contributions of these neglected Aβ variants to microglia activation. First, the impact of Aβ receptors, signaling cascades, scavenger mechanisms, and genetic variations on the physiological responses towards various Aβ species is described. Furthermore, we discuss the importance of different types of amyloid precursor protein processing for the generation of these Aβ variants in microglia, astrocytes, oligodendrocytes, and neurons, and highlight how alterations in secondary structures and oligomerization affect Aβ neurotoxicity. In sum, the data indicate that gene polymorphisms in Aβ-driven signaling pathways in combination with the production and activity of different Aβ variants might be crucial factors for the initiation and progression of different forms of AD. A deeper assessment of their interplay with glial cells may pave the way towards novel therapeutic strategies for individualized medicine.
Collapse
Affiliation(s)
- Lukas Busch
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibruecken, Germany
| | - Simone Eggert
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, D-37075 Goettingen, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Centre of the Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Bernd Bufe
- Department of Informatics and Microsystems Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibruecken, Germany
| |
Collapse
|
14
|
Huwait EA, Baghallab IM, Glabe CG, Abulnaja KO, Kumosani TA, Moselhy SS. Identification of amyloid antibodies for Alzheimer disease - immunotherapy. Arch Physiol Biochem 2022; 128:1275-1282. [PMID: 32449861 DOI: 10.1080/13813455.2020.1767147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The current study identified the specific antibodies that recognise amyloid protein for Alzheimer disease - immunotherapy. The immune-selection of random sequences from a phage display library and sequencing to obtain the random 12 amino acids peptide library for each antibody, and then we analysed these peptides for unique and common sequences, relation to Aβ42 sequence and shape and pattern of the amino acid reaction to the antibody to predict the epitopes. Data obtained for 4G8 showed that, the sequence segment related to the putative epitope of 4G8 was LVFFAED. Nine of the ten top sequences contain the sequence RHD corresponding to the Aβ sequence from residues 5-7. Peptide 7 has the sequence IRYDTGSYHIH, which has a RYD. It was concluded that, 4G8 and 6E10 can tolerate the binding the sequences that explain it is able to recognise amyloid aggregates.
Collapse
Affiliation(s)
- Etimad A Huwait
- Biochemistry Department, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Ibtisam M Baghallab
- Biochemistry Department, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
| | - Charles G Glabe
- Biochemistry Department, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Khalid O Abulnaja
- Biochemistry Department, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, KAU, Jeddah, Saudi Arabia
- Bioactive Natural Products Research Group, KAU, Jeddah, Saudi Arabia
| | - Taha A Kumosani
- Biochemistry Department, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, KAU, Jeddah, Saudi Arabia
- Production of Bio-Products for Industrial Applications Research Group, KAU, Jeddah, Saudi Arabia
| | - Said S Moselhy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
15
|
Regulation of the Soluble Amyloid Precursor Protein α (sAPPα) Levels by Acetylcholinesterase and Brain-Derived Neurotrophic Factor in Lung Cancer Cell Media. Int J Mol Sci 2022; 23:ijms231810746. [PMID: 36142659 PMCID: PMC9500850 DOI: 10.3390/ijms231810746] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/10/2022] [Indexed: 11/17/2022] Open
Abstract
In comparing two human lung cancer cells, we previously found lower levels of acetylcholinesterase (AChE) and intact amyloid-β40/42 (Aβ), and higher levels of mature brain-derived neurotrophic factor (mBDNF) in the media of H1299 cells as compared to A549 cell media. In this study, we hypothesized that the levels of soluble amyloid precursor protein α (sAPPα) are regulated by AChE and mBDNF in A549 and H1299 cell media. The levels of sAPPα were higher in the media of H1299 cells. Knockdown of AChE led to increased sAPPα and mBDNF levels and correlated with decreased levels of intact Aβ40/42 in A549 cell media. AChE and mBDNF had opposite effects on the levels of Aβ and sAPPα and were found to operate through a mechanism involving α-secretase activity. Treatment with AChE decreased sAPPα levels and simultaneously increased the levels of intact Aβ40/42 suggesting a role of the protein in shifting APP processing away from the non-amyloidogenic pathway and toward the amyloidogenic pathway, whereas treatment with mBDNF led to opposite effects on those levels. We also show that the levels of sAPPα are regulated by protein kinase C (PKC), extracellular signal-regulated kinase (ERK)1/2, phosphoinositide 3 Kinase (PI3K), but not by protein kinase A (PKA).
Collapse
|
16
|
Fixemer S, Ameli C, Hammer G, Salamanca L, Uriarte Huarte O, Schwartz C, Gérardy JJ, Mechawar N, Skupin A, Mittelbronn M, Bouvier DS. Microglia phenotypes are associated with subregional patterns of concomitant tau, amyloid-β and α-synuclein pathologies in the hippocampus of patients with Alzheimer's disease and dementia with Lewy bodies. Acta Neuropathol Commun 2022; 10:36. [PMID: 35296366 PMCID: PMC8925098 DOI: 10.1186/s40478-022-01342-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/26/2022] Open
Abstract
The cellular alterations of the hippocampus lead to memory decline, a shared symptom between Alzheimer’s disease (AD) and dementia with Lewy Bodies (DLB) patients. However, the subregional deterioration pattern of the hippocampus differs between AD and DLB with the CA1 subfield being more severely affected in AD. The activation of microglia, the brain immune cells, could play a role in its selective volume loss. How subregional microglia populations vary within AD or DLB and across these conditions remains poorly understood. Furthermore, how the nature of the hippocampal local pathological imprint is associated with microglia responses needs to be elucidated. To this purpose, we employed an automated pipeline for analysis of 3D confocal microscopy images to assess CA1, CA3 and DG/CA4 subfields microglia responses in post-mortem hippocampal samples from late-onset AD (n = 10), DLB (n = 8) and age-matched control (CTL) (n = 11) individuals. In parallel, we performed volumetric analyses of hyperphosphorylated tau (pTau), amyloid-β (Aβ) and phosphorylated α-synuclein (pSyn) loads. For each of the 32,447 extracted microglia, 16 morphological features were measured to classify them into seven distinct morphological clusters. Our results show similar alterations of microglial morphological features and clusters in AD and DLB, but with more prominent changes in AD. We identified two distinct microglia clusters enriched in disease conditions and particularly increased in CA1 and DG/CA4 of AD and CA3 of DLB. Our study confirms frequent concomitance of pTau, Aβ and pSyn loads across AD and DLB but reveals a specific subregional pattern for each type of pathology, along with a generally increased severity in AD. Furthermore, pTau and pSyn loads were highly correlated across subregions and conditions. We uncovered tight associations between microglial changes and the subfield pathological imprint. Our findings suggest that combinations and severity of subregional pTau, Aβ and pSyn pathologies transform local microglia phenotypic composition in the hippocampus. The high burdens of pTau and pSyn associated with increased microglial alterations could be a factor in CA1 vulnerability in AD.
Collapse
|
17
|
Tournier BB, Tsartsalis S, Ceyzériat K, Fraser BH, Grégoire MC, Kövari E, Millet P. Astrocytic TSPO Upregulation Appears Before Microglial TSPO in Alzheimer's Disease. J Alzheimers Dis 2021; 77:1043-1056. [PMID: 32804124 PMCID: PMC7683091 DOI: 10.3233/jad-200136] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background: In vivo PET/SPECT imaging of neuroinflammation is primarily based on the estimation of the 18 kDa-translocator-protein (TSPO). However, TSPO is expressed by different cell types which complicates the interpretation. Objective: The present study evaluates the cellular origin of TSPO alterations in Alzheimer’s disease (AD). Methods: The TSPO cell origin was evaluated by combining radioactive imaging approaches using the TSPO radiotracer [125I]CLINDE and fluorescence-activated cell sorting, in a rat model of AD (TgF344-AD) and in AD subjects. Results: In the hippocampus of TgF344-AD rats, TSPO overexpression not only concerns glial cells but the increase is visible at 12 and 24 months in astrocytes and only at 24 months in microglia. In the temporal cortex of AD subjects, TSPO upregulation involved only glial cells. However, the mechanism of this upregulation appears different with an increase in the number of TSPO binding sites per cell without cell proliferation in the rat, and a microglial cell population expansion with a constant number of binding sites per cell in human AD. Conclusion: These data indicate an earlier astrocyte intervention than microglia and that TSPO in AD probably is an exclusive marker of glial activity without interference from other TSPO-expressing cells. This observation indicates that the interpretation of TSPO imaging depends on the stage of the pathology, and highlights the particular role of astrocytes.
Collapse
Affiliation(s)
- Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland
| | - Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland
| | - Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland.,Division of Nuclear medicine, University Hospitals of Geneva, Switzerland
| | - Ben H Fraser
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Sydney, NSW, Australia
| | - Marie-Claude Grégoire
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Sydney, NSW, Australia
| | - Enikö Kövari
- Division of Geriatric Psychiatry, Department of Mental Health and Psychiatry, University Hospitals of Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Switzerland
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Switzerland
| |
Collapse
|
18
|
Is Cerebral Amyloid-β Deposition Related to Post-stroke Cognitive Impairment? Transl Stroke Res 2021; 12:946-957. [PMID: 34195928 DOI: 10.1007/s12975-021-00921-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 01/20/2023]
Abstract
Approximately two-thirds of ischemic stroke patients suffer from different levels of post-stroke cognitive impairment (PSCI), but the underlying mechanisms of PSCI remain unclear. Cerebral amyloid-β (Aβ) deposition, a pathological hallmark of Alzheimer's disease, has been discovered in the brains of stroke patients in some autopsy studies. However, less is known about the role of Aβ pathology in the development of PSCI. It is hypothesized that cerebral ischemic injury may lead to neurotoxic Aβ accumulation in the brain, which further induces secondary neurodegeneration and progressive cognitive decline after stroke onset. In this review, we summarized available evidence from pre-clinical and clinical studies relevant to the aforementioned hypothesis. We found inconsistency in the results obtained from studies in rodents, nonhuman primates, and stroke patients. Moreover, the causal relationship between post-stroke cerebral Aβ deposition and PSCI has been uncertain and controversial. Taken together, evidence supporting the hypothesis that brain ischemia induces cerebral Aβ deposition has been insufficient so far. And, there is still no consensus regarding the contribution of cerebral amyloid pathology to PSCI. Other non-amyloid neurodegenerative mechanisms might be involved and remain to be fully elucidated.
Collapse
|
19
|
Maciejewska K, Czarnecka K, Szymański P. A review of the mechanisms underlying selected comorbidities in Alzheimer's disease. Pharmacol Rep 2021; 73:1565-1581. [PMID: 34121170 PMCID: PMC8599320 DOI: 10.1007/s43440-021-00293-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder of the central nervous system (CNS) leading to mental deterioration and devastation, and eventually a fatal outcome. AD affects mostly the elderly. AD is frequently accompanied by hypercholesterolemia, hypertension, atherosclerosis, and diabetes mellitus, and these are significant risk factors of AD. Other conditions triggered by the progression of AD include psychosis, sleep disorders, epilepsy, and depression. One important comorbidity is Down’s syndrome, which directly contributes to the severity and rapid progression of AD. The development of new therapeutic strategies for AD includes the repurposing of drugs currently used for the treatment of comorbidities. A better understanding of the influence of comorbidities on the pathogenesis of AD, and the medications used in its treatment, might allow better control of disease progression, and more effective pharmacotherapy.
Collapse
Affiliation(s)
- Karolina Maciejewska
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Kamila Czarnecka
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St, 01-163, Warsaw, Poland
| | - Paweł Szymański
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland.
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St, 01-163, Warsaw, Poland.
| |
Collapse
|
20
|
Guo L, Ravindran N, Shamsher E, Hill D, Cordeiro MF. Retinal Changes in Transgenic Mouse Models of Alzheimer's Disease. Curr Alzheimer Res 2021; 18:89-102. [PMID: 33855942 DOI: 10.2174/1567205018666210414113634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/09/2021] [Accepted: 04/05/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, the most common form of dementia. AD is characterised by amyloid-β (Aβ) plaques and neurofibrillary tangles (NFT) in the brain, in association with neuronal loss and synaptic failure, causing cognitive deficits. Accurate and early diagnosis is currently unavailable in lifespan, hampering early intervention of potential new treatments. Visual deficits have been well documented in AD patients, and the pathological changes identified in the brain are also believed to be found in the retina, an integral part of the central nervous system. Retinal changes can be detected by real-time non-invasive imaging, due to the transparent nature of the ocular media, potentially allowing an earlier diagnosis as well as monitoring disease progression and treatment outcome. Animal models are essential for AD research, and this review has a focus on retinal changes in various transgenic AD mouse models with retinal imaging and immunohistochemical analysis as well as therapeutic effects in those models. We also discuss the limitations of transgenic AD models in clinical translations.
Collapse
Affiliation(s)
- Li Guo
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Nivedita Ravindran
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Ehtesham Shamsher
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Daniel Hill
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - M Francesca Cordeiro
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
21
|
Dorandish S, Williams A, Atali S, Sendo S, Price D, Thompson C, Guthrie J, Heyl D, Evans HG. Regulation of amyloid-β levels by matrix metalloproteinase-2/9 (MMP2/9) in the media of lung cancer cells. Sci Rep 2021; 11:9708. [PMID: 33958632 PMCID: PMC8102533 DOI: 10.1038/s41598-021-88574-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
In this study, we set out to identify regulators of intact amyloid-β40/42 (Aβ) levels in A549 (p53 wild-type) and H1299 (p53-null) lung cancer cell media. Higher Aβ levels were detected in the media of A549 than H1299 cells without or with treatment with 4-methylumbelliferone (4-MU) and/or the anti-CD44 antibody (5F12). Using inhibitors, we found that PI3K, AKT, and NFκB are likely involved in regulating Aβ levels in the media. However, increased Aβ levels that more closely resembled those found upon 4-MU co-treatment resulted from MMP2/9 inhibition, suggesting that MMP2/9 maybe the main contributors to regulation of Aβ levels in the media. Differences in Aβ levels might be accounted for, in part, by p53 since blocking p53 function in A549 cells resulted in decreased Aβ levels, increased MMP2/9 levels, increased PI3K/AKT activities and the phospho/total NFκB ratio. Using siRNA targeted against MMP2 or MMP9, we found increased Aβ levels in the media, however, MMP2 knockdown led to Aβ levels closely mimicking those detected by co-treatment with 4-MU. Cell viability or apoptosis upon treatment with either MMP2 or MMP9 siRNA along with Aβ immunodepletion, showed that MMP2 is the predominant regulator of the cytotoxic effects induced by Aβ in lung cancer cells.
Collapse
Affiliation(s)
- Sadaf Dorandish
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Asana Williams
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Sarah Atali
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Sophia Sendo
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Deanna Price
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Colton Thompson
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Jeffrey Guthrie
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Deborah Heyl
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Hedeel Guy Evans
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI, 48197, USA.
| |
Collapse
|
22
|
Hunter S, Brayne C. Amyloid in the ageing brain: New frameworks and perspectives. AGING BRAIN 2021; 1:100008. [PMID: 36911501 PMCID: PMC9997141 DOI: 10.1016/j.nbas.2021.100008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 01/23/2023] Open
Affiliation(s)
- Sally Hunter
- Cambridge Public Health, University of Cambridge School of Clinical Medicine, Forvie Site, Cambridge Biomedical Campus, Cambridge CB2 0SR, United Kingdom
| | - Carol Brayne
- Cambridge Public Health, University of Cambridge School of Clinical Medicine, Forvie Site, Cambridge Biomedical Campus, Cambridge CB2 0SR, United Kingdom
| |
Collapse
|
23
|
Tiiman A, Jelić V, Jarvet J, Järemo P, Bogdanović N, Rigler R, Terenius L, Gräslund A, Vukojević V. Amyloidogenic Nanoplaques in Blood Serum of Patients with Alzheimer's Disease Revealed by Time-Resolved Thioflavin T Fluorescence Intensity Fluctuation Analysis. J Alzheimers Dis 2020; 68:571-582. [PMID: 30814355 PMCID: PMC6484272 DOI: 10.3233/jad-181144] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Biomarkers are central to current research on molecular mechanisms underlying Alzheimer's disease (AD). Their further development is of paramount importance for understanding pathophysiological processes that eventually lead to disease onset. Biomarkers are also crucial for early disease detection, before clinical manifestation, and for development of new disease modifying therapies. OBJECTIVE The overall aim of this work is to develop a minimally invasive method for fast, ultra-sensitive and cost-effective detection of structurally modified peptide/protein self-assemblies in the peripheral blood and in other biological fluids. Specifically, we focus here on using this method to detect structured amyloidogenic oligomeric aggregates in the blood serum of apparently healthy individuals and patients in early AD stage, and measure their concentration and size. METHODS Time-resolved detection of Thioflavin T (ThT) fluorescence intensity fluctuations in a sub-femtoliter observation volume element was used to identify in blood serum ThT-active structured amyloidogenic oligomeric aggregates, hereafter called nanoplaques, and measure with single-particle sensitivity their concentration and size. RESULTS The concentration and size of structured amyloidogenic nanoplaques are significantly higher in the blood serum of individuals diagnosed with AD than in control subjects. CONCLUSION A new method with the ultimate, single-particle sensitivity was successfully developed. The proposed approach neither relies on the use of immune-based probes, nor on the use of radiotracers, signal-amplification or protein separation techniques, and provides a minimally invasive test for fast and cost-effective early determination of structurally modified peptides/proteins in the peripheral blood, as shown here, but also in other biological fluids.
Collapse
Affiliation(s)
- Ann Tiiman
- Department of Clinical Neuroscience (CNS), Center for Molecular Medicine CMM L8:01, Karolinska Institutet, Stockholm, Sweden
| | - Vesna Jelić
- Department of Neurobiology, Care Sciences and Society (NVS), Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, Stockholm, Sweden.,The National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Petter Järemo
- Department of Internal Medicine, The Vrinnevi Hospital, Norrköping, Sweden
| | - Nenad Bogdanović
- Department of Neurobiology, Care Sciences and Society (NVS), Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden.,Department of Geriatric Medicine, University of Oslo, Oslo, Norway
| | - Rudolf Rigler
- Department of Medical Biochemistry and Biophysics (MBB), Karolinska Institutet, Stockholm, Sweden
| | - Lars Terenius
- Department of Clinical Neuroscience (CNS), Center for Molecular Medicine CMM L8:01, Karolinska Institutet, Stockholm, Sweden
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, Stockholm, Sweden
| | - Vladana Vukojević
- Department of Clinical Neuroscience (CNS), Center for Molecular Medicine CMM L8:01, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Bergkvist L, Du Z, Elovsson G, Appelqvist H, Itzhaki LS, Kumita JR, Kågedal K, Brorsson AC. Mapping pathogenic processes contributing to neurodegeneration in Drosophila models of Alzheimer's disease. FEBS Open Bio 2020; 10:338-350. [PMID: 31823504 PMCID: PMC7050262 DOI: 10.1002/2211-5463.12773] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/21/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, affecting millions of people and currently lacking available disease‐modifying treatments. Appropriate disease models are necessary to investigate disease mechanisms and potential treatments. Drosophila melanogaster models of AD include the Aβ fly model and the AβPP‐BACE1 fly model. In the Aβ fly model, the Aβ peptide is fused to a secretion sequence and directly overexpressed. In the AβPP‐BACE1 model, human AβPP and human BACE1 are expressed in the fly, resulting in in vivo production of Aβ peptides and other AβPP cleavage products. Although these two models have been used for almost two decades, the underlying mechanisms resulting in neurodegeneration are not yet clearly understood. In this study, we have characterized toxic mechanisms in these two AD fly models. We detected neuronal cell death and increased protein carbonylation (indicative of oxidative stress) in both AD fly models. In the Aβ fly model, this correlates with high Aβ1–42 levels and down‐regulation of the levels of mRNA encoding lysosomal‐associated membrane protein 1, lamp1 (a lysosomal marker), while in the AβPP‐BACE1 fly model, neuronal cell death correlates with low Aβ1–42 levels, up‐regulation of lamp1 mRNA levels and increased levels of C‐terminal fragments. In addition, a significant amount of AβPP/Aβ antibody (4G8)‐positive species, located close to the endosomal marker rab5, was detected in the AβPP‐BACE1 model. Taken together, this study highlights the similarities and differences in the toxic mechanisms which result in neuronal death in two different AD fly models. Such information is important to consider when utilizing these models to study AD pathogenesis or screening for potential treatments.
Collapse
Affiliation(s)
- Liza Bergkvist
- Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, Sweden
| | - Zhen Du
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, UK.,Department of Pharmacology, University of Cambridge, UK
| | - Greta Elovsson
- Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, Sweden
| | - Hanna Appelqvist
- Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, Sweden.,Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Sweden
| | | | - Janet R Kumita
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, UK
| | - Katarina Kågedal
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Sweden
| | - Ann-Christin Brorsson
- Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, Sweden
| |
Collapse
|
25
|
Liu Y, Wei W, Baazaoui N, Liu F, Iqbal K. Inhibition of AMD-Like Pathology With a Neurotrophic Compound in Aged Rats and 3xTg-AD Mice. Front Aging Neurosci 2019; 11:309. [PMID: 31803044 PMCID: PMC6877482 DOI: 10.3389/fnagi.2019.00309] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 10/28/2019] [Indexed: 11/17/2022] Open
Abstract
Age-associated macular degeneration (AMD), which leads to loss of vision at its end stage, is one of the most common neurodegenerative diseases among the elderly. However, to date, no effective drug therapy is available for the prevention of AMD. Here, we report the occurrence of AMD pathology and its prevention by chronic treatment with the neurotrophic peptidergic compound P021, in aged rats and 3xTg-AD mice. We found photoreceptor degeneration, lipofuscin granules, vacuoles, and atrophy in retinal pigment epithelium (RPE) as well as Bruch’s membrane (BM) thickening; in aged rats, we even found rosette-like structure formation. Microgliosis and astrogliosis were observed in different retinal layers. In addition, we also found that total tau, phosphorylated tau, Aβ/APP, and VEGF were widely distributed in the sub-retina of aged rats and 3xTg mice. Importantly, chronic treatment with P021 for 3 months in rats and for 18 months in 3xTg mice ameliorated the pathological changes above. These findings indicate the therapeutic potential of P021 for prevention and treatment of AMD and retinal changes associated with aging and Alzheimer’s disease.
Collapse
Affiliation(s)
- Yinghua Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States.,Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Molecular Clinical Pharmacology, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wei Wei
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States.,Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, China
| | - Narjes Baazaoui
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| |
Collapse
|
26
|
Joseph DJ, Liu C, Peng J, Liang G, Wei H. Isoflurane mediated neuropathological and cognitive impairments in the triple transgenic Alzheimer's mouse model are associated with hippocampal synaptic deficits in an age-dependent manner. PLoS One 2019; 14:e0223509. [PMID: 31600350 PMCID: PMC6786564 DOI: 10.1371/journal.pone.0223509] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/22/2019] [Indexed: 12/17/2022] Open
Abstract
Many in vivo studies suggest that inhalational anesthetics can accelerate or prevent the progression of neuropathology and cognitive impairments in Alzheimer Disease (AD), but the synaptic mechanisms mediating these ambiguous effects are unclear. Here, we show that repeated exposures of neonatal and old triple transgenic AD (3xTg) and non-transgenic (NonTg) mice to isoflurane (Iso) distinctly increased neurodegeneration as measured by S100β levels, intracellular Aβ, Tau oligomerization, and apoptotic markers. Spatial cognition measured by reference and working memory testing in the Morris Water Maze (MWM) were altered in young NonTg and 3xTg. Field recordings in the cornu ammonis 1 (CA1) hippocampus showed that neonatal control 3xTg mice exhibited hypo-excitable synaptic transmission, reduced paired-pulse facilitation (PPF), and normal long-term potentiation (LTP) compared to NonTg controls. By contrast, the old control 3xTg mice exhibited hyper-excitable synaptic transmission, enhanced PPF, and unstable LTP compared to NonTg controls. Repeated Iso exposures reduced synaptic transmission and PPF in neonatal NonTg and old 3xTg mice. LTP was normalized in old 3xTg mice, but reduced in neonates. By contrast, LTP was reduced in old but not neonatal NonTg mice. Our results indicate that Iso-mediated neuropathologic and cognitive defects in AD mice are associated with synaptic pathologies in an age-dependent manner. Based on these findings, the extent of this association with age and, possibly, treatment paradigms warrant further study.
Collapse
Affiliation(s)
- Donald J. Joseph
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Chunxia Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Jun Peng
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Anesthesiology, sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ge Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
27
|
Ouyang F, Chen X, Chen Y, Liang J, Chen Y, Lu T, Huang W, Zeng J. Neuronal loss without amyloid-β deposits in the thalamus and hippocampus in the late period after middle cerebral artery occlusion in cynomolgus monkeys. Brain Pathol 2019; 30:165-178. [PMID: 31278793 DOI: 10.1111/bpa.12764] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/01/2019] [Indexed: 11/30/2022] Open
Abstract
Conflicting evidence exists regarding whether focal cerebral infarction contributes to cerebral amyloid-β (Aβ) deposition, as observed in Alzheimer's disease. In this study, we aimed to evaluate the presence of Aβ deposits in the ipsilateral thalamus and hippocampus 12 months post-stroke in non-human primates, whose brains are structurally and functionally similar to that of humans. Four young male cynomolgus monkeys were subjected to unilateral permanent middle cerebral artery occlusion (MCAO), and another four sham-operated monkeys served as controls. All monkeys underwent magnetic resonance imaging examination on post-operative day 7 to assess the location and size of the infarction. The numbers of neurons, astrocytes, microglia and the Aβ load in the non-affected thalamus and hippocampus ipsilaterally remote from infarct foci were examined immunohistochemically at sacrifice 12 months after operation. Thioflavin S and Congo Red stainings were used to identify amyloid deposits. Multiple Aβ antibodies recognizing both the N-terminal and C-terminal epitopes of Aβ peptides were used to avoid antibody cross-reactivity. Aβ levels in cerebrospinal fluid (CSF) and plasma were examined using enzyme-linked immunosorbent assay. The initial infarct was restricted to the left temporal, parietal, insular cortex and the subcortical white matter, while the thalamus and hippocampus remained intact. Of note, there were fewer neurons and more glia in the ipsilateral thalamus and hippocampus in the MCAO group at 12 months post-stroke compared to the control group (all P < 0.05). However, there was no sign of extracellular Aβ plaques in the thalamus or hippocampus. No statistically significant difference was found in CSF or plasma levels of Aβ40 , Aβ42 or the Aβ40 /Aβ42 ratio between the two groups (P > 0.05). These results suggest that significant secondary neuronal loss and reactive gliosis occur in the non-affected thalamus and hippocampus without Aβ deposits in the late period after MCAO in non-human primates.
Collapse
Affiliation(s)
- Fubing Ouyang
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinran Chen
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yonghong Chen
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiahui Liang
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yicong Chen
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Lu
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weixian Huang
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinsheng Zeng
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Mouchard A, Boutonnet MC, Mazzocco C, Biendon N, Macrez N. ApoE-fragment/Aβ heteromers in the brain of patients with Alzheimer's disease. Sci Rep 2019; 9:3989. [PMID: 30850702 PMCID: PMC6408522 DOI: 10.1038/s41598-019-40438-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/13/2019] [Indexed: 11/09/2022] Open
Abstract
Identification of endogenous pathological amyloid β peptides (Aβ) forms in the brains of patients with Alzheimer’s disease (AD) is still unclear. In healthy brain, Aβ can associate with Apolipoprotein E (ApoE) which is involved in its metabolism and clearance. In the brain of patients with AD, ApoE is cleaved and produces ApoE fragments. We studied the forms of Aβ and their interaction with the ApoE fragments in post-mortem brains from control and AD patients by western blots and co-immunoprecipitation. Three Aβ-containing peptides and three ApoE fragments were specifically found in the brain of AD patients. Co-immunoprecipitations showed that ApoE fragments and Aβ1–42 peptides are co-partners in heteromers of 18 and 16 kDa while ApoE-fragments and Aβ peptides of 12 kDa did not interact with each other. Formation of the 18 kDa ApoE-fragment/Aβ heteromers is specifically increased in ApoE4 carriers and is a strong brain marker of AD while 16 kDa ApoE-fragment/Aβ and Aβ 12 kDa correlate to memory deficit. These data show that in patients with AD, ApoE fragmentation generates peptides that trap Aβ in the brain. Inhibiting the fragmentation or targeting ApoE fragments could be exploited to define strategies to detect or reverse AD.
Collapse
Affiliation(s)
- Amandine Mouchard
- Bordeaux University, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France
| | - Marie-Charlotte Boutonnet
- Bordeaux University, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France
| | - Claire Mazzocco
- Bordeaux University, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France
| | - Nathalie Biendon
- Bordeaux University, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France
| | - Nathalie Macrez
- Bordeaux University, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France. .,CNRS, Institut des Maladies Neurodégénératives, UMR, 5293, Bordeaux, France.
| | | |
Collapse
|
29
|
den Haan J, Morrema THJ, Verbraak FD, de Boer JF, Scheltens P, Rozemuller AJ, Bergen AAB, Bouwman FH, Hoozemans JJ. Amyloid-beta and phosphorylated tau in post-mortem Alzheimer's disease retinas. Acta Neuropathol Commun 2018; 6:147. [PMID: 30593285 PMCID: PMC6309096 DOI: 10.1186/s40478-018-0650-x] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 01/02/2023] Open
Abstract
In-vivo labeling of retinal amyloid-beta(Aβ) and tau has potential as non-invasive biomarker for Alzheimer's disease (AD). However, literature on the presence of Aβ and phosphorylated tau (pTau) in AD retinas is inconclusive. We therefore assessed the presence of Aβ and pTau in post-mortem retinas in 6 AD and 6 control cases who donated brains and eyes to the Netherlands Brain Bank. Neuropathological diagnosis of AD was made according to NIA-AA criteria. Formalin fixed retinas were dissected in quadrants and cross-sections of medial and superior retinas were made. Immuno-histochemical stainings were performed for Aβ, amyloid precursor protein (APP) and pTau. To assess translation to an in-vivo set up using curcumin as labelling fluorophore, co-stainings with curcumin were performed. No typical Aβ-plaques and neurofibrillary tangles, like in the cerebral cortex, were observed in AD retinas. A diffuse immunoreactive signal for pTau was increased in the inner and outer plexiform layers of the retina in AD cases compared to control cases with absence of cerebral amyloid pathology. Immunostaining with anti-Aβ and anti-APP antibodies yielded signal in ganglion cells, amacrine cells, horizontal cells and Müller cells in both control and AD cases. We observed small extracellular deposits positive for anti-Aβ antibodies 12F4 and 6E10 and negative for 4G8 and curcumin. A subset of these deposits could be characterized as corpora amylacea. In conclusion we found that retinal manifestations of AD pathology appear to be different compared to cerebral AD pathology. Using a qualitative cross-sectional approach, we did not find Aβ/APP related differences in the retina between AD and control subjects. In contrast, tau related changes were found to be present in cases with cerebral AD pathology, suggesting retinal tau as a potential biomarker for AD.
Collapse
Affiliation(s)
- Jurre den Haan
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Mailbox 7057, Amsterdam, 1007 MB, The Netherlands.
| | - Tjado H J Morrema
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Frank D Verbraak
- Ophthalmology Department, VU University Medical Center, Amsterdam, The Netherlands
| | - Johannes F de Boer
- Department of Physics, Bio Laser Lab Amsterdam, VU University, Amsterdam, The Netherlands
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Mailbox 7057, Amsterdam, 1007 MB, The Netherlands
| | - Annemieke J Rozemuller
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Arthur A B Bergen
- Departments of Clinical genetics and Ophthalmology, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN-KNAW), Amsterdam, The Netherlands
| | - Femke H Bouwman
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Mailbox 7057, Amsterdam, 1007 MB, The Netherlands
| | - Jeroen J Hoozemans
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Hunter S, Smailagic N, Brayne C. Aβ and the dementia syndrome: Simple versus complex perspectives. Eur J Clin Invest 2018; 48:e13025. [PMID: 30246866 DOI: 10.1111/eci.13025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 08/15/2018] [Accepted: 09/06/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The amyloid cascade hypothesis (ACH) has dominated strategy in dementia research for decades despite evidence of its limitations including known heterogeneity of the dementia syndrome in the population and the narrow focus on a single molecule - the amyloid beta protein (Aβ) as causal for all Alzheimer-type dementia. Other hypotheses relevant to Aβ are the presenilin (PS) hypothesis (PSH) relating to the involvement of PS in the generation of Aβ, and the amyloid precursor protein (APP) matrix approach (AMA), relating to the complex and dynamic breakdown of APP, from which Aβ derives. MATERIALS AND METHODS In this article we explore perspectives relating to complex disorders occurring mainly in older populations through a detailed case study of the role of Aβ in AD. RESULTS Scrutiny of the evidence generated so far reveals and a lack of understanding of the wider APP proteolytic system and how narrow research into the dementia syndrome has been to date. Confounding factors add significant limitations to the understanding of the current evidence base. CONCLUSIONS A better characterisation of the entire APP proteolytic system in the human brain is urgently required to place Aβ in its complex physiological context. From a molecular perspective, a combination of the alternative hypotheses, the PSH and the AMA may better describe the complexity of the APP proteolytic system leading to new therapeutic approaches. The reductionist approach is widespread throughout biomedical research and this example highlights how neglect of complexity can undermine investigations of complex disorders, particularly those arising in the oldest in our populations.
Collapse
Affiliation(s)
- Sally Hunter
- Department of Public Health and Primary Care, Institute of Public Health, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Nadja Smailagic
- Department of Public Health and Primary Care, Institute of Public Health, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Department of Public Health and Primary Care, Institute of Public Health, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
31
|
Zhang Y, Huai Y, Zhang X, Song C, Cai J, Zhang Y. The Mode of Action of an Anti-Oligomeric Amyloid β-Protein Antibody Affects its Protective Efficacy. Neurotox Res 2018; 35:304-317. [PMID: 30229545 DOI: 10.1007/s12640-018-9955-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 12/22/2022]
Abstract
The process of developing antibody drugs for Alzheimer's disease therapy has been both long and difficult; however, recent advances suggest that antibodies against neurotoxic Αβ42 can suppress the progression of AD, especially on its early stage. Here, we obtained and characterized a novel anti-oligomeric Aβ42 aggregate scFv antibody, HT7, which could induce the significant disaggregation of Aβ42 aggregates through the release of stable and non-cytotoxic hexameric complexes that were composed of three scFv HT7s and one Aβ42 trimer, the latter being found to serve as the assembled subunit within larger Aβ42 aggregates in addition to existing freely between the cells. The docking model of the scFv HT7-Aβ42 complex revealed that only the N-terminal peptide of the Aβ42 molecule was bound into the groove between the VH and VL domains of scFv HT7. Thus, it was suggested that the hydrophobic interaction between the C-terminal peptides of Aβ42 molecules maintained the stability of the Aβ42 trimers or the Aβ42 trimer subunits. The saturation of Aβ42 trimer subunits by scFv HT7 and the subsequent dissociation of the scFv HT7-saturated Aβ42 trimer subunits from larger Aβ42 aggregates constituted the primary mechanisms underlying the high efficacy of scFv HT7. Our findings revealed that it was not sufficient for an anti-oligomeric Aβ42 antibody to exhibit high specificity and high affinity to the oligomeric Aβ42 aggregates in order to promote Aβ42 aggregate clearance and neutralize their cytotoxic effects. Here, for the first time, we proposed a "post-saturation dissociation" mechanism of Aβ42 oligomeric subunits for effective anti-Aβ42 antibodies.
Collapse
Affiliation(s)
- Yunlong Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China
| | - Yangyang Huai
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China
| | - Xiaoning Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China
| | - Chuli Song
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China
| | - Jing Cai
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China.,China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yingjiu Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China. .,School of Life Science, Jilin University, Changchun, China.
| |
Collapse
|
32
|
Shi Y, Huang W, Wang Y, Zhang R, Hou L, Xu J, Qiu Z, Xie Q, Chen H, Zhang Y, Wang H. Bis(9)-(-)-Meptazinol, a novel dual-binding AChE inhibitor, rescues cognitive deficits and pathological changes in APP/PS1 transgenic mice. Transl Neurodegener 2018; 7:21. [PMID: 30237878 PMCID: PMC6142624 DOI: 10.1186/s40035-018-0126-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/20/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative brain disorder, which is the most common form of dementia. Intensive efforts have been made to find effective and safe treatment against AD. Acetylcholinesterase inhibitors (AChEIs) have been widely used for the treatment of mild to moderate AD. In this study, we investigated the effect of Bis(9)-(-)-Meptazinol (B9M), a novel potential dual-binding acetylcholinesterase (AChE) inhibitor, on learning and memory abilities, as well as the underlying mechanism in the APP/PS1 mouse model of AD. METHODS B9M (0.1 μg/kg, 0.3 μg/kg, and 1 μg/kg) was administered by subcutaneous injection into eight-month-old APP/PS1 transgenic mice for four weeks. Morris water maze, nest-building and novel object recognition were used to examine learning and memory ability. Aβ levels and Aβ plaque were evaluated by ELISA and immunochemistry. RESULTS Our results showed that chronic treatment with B9M significantly improved the cognitive function of APP/PS1 transgenic mice in the Morris water maze test, nest-building test and novel object recognition test. Moreover, B9M improved cognitive deficits in APP/PS1 mice by a mechanism that may be associated with its inhibition of the AChE activity, Aβ plaque burden, levels of Aβ and the consequent activation of astrocytes and microglia in the brain of APP/PS1 transgenic mice. Most of important, the most effective dose of B9M in the present study is 1 μg/kg, which is one thousand of the dosage of Donepezil acted as the control treatment. Furthermore, B9M reduced Aβ plaque burden better than Donepezil. CONCLUSION These results indicate that B9M appears to have potential as an effective AChE inhibitor for the treatment of AD with symptom-relieving and disease-modifying properties.
Collapse
Affiliation(s)
- Yuhuan Shi
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai, 200025 People’s Republic of China
| | - Wanying Huang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai, 200025 People’s Republic of China
| | - Yu Wang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai, 200025 People’s Republic of China
| | - Rui Zhang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai, 200025 People’s Republic of China
| | - Lina Hou
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai, 200025 People’s Republic of China
| | - Jianrong Xu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai, 200025 People’s Republic of China
| | - Zhuibai Qiu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, People’s Republic of China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, People’s Republic of China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai, 200025 People’s Republic of China
| | - Yongfang Zhang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai, 200025 People’s Republic of China
| | - Hao Wang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai, 200025 People’s Republic of China
| |
Collapse
|
33
|
Herzer S, Hagan C, von Gerichten J, Dieterle V, Munteanu B, Sandhoff R, Hopf C, Nordström V. Deletion of Specific Sphingolipids in Distinct Neurons Improves Spatial Memory in a Mouse Model of Alzheimer's Disease. Front Mol Neurosci 2018; 11:206. [PMID: 29973867 PMCID: PMC6019486 DOI: 10.3389/fnmol.2018.00206] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/25/2018] [Indexed: 01/25/2023] Open
Abstract
Alzheimer’s disease (AD) is characterized by progressive neurodegeneration and a concomitant loss of synapses and cognitive abilities. Recently, we have proposed that an alteration of neuronal membrane lipid microdomains increases neuronal resistance toward amyloid-β stress in cultured neurons and protects from neurodegeneration in a mouse model of AD. Lipid microdomains are highly enriched in a specific subclass of glycosphingolipids, termed gangliosides. The enzyme glucosylceramide synthase (GCS) catalyzes the rate-limiting step in the biosynthesis of these gangliosides. The present work now demonstrates that genetic GCS deletion in subsets of adult forebrain neurons significantly improves the spatial memory and counteracts the loss of dendritic spines in the hippocampal dentate gyrus of 5x familial AD mice (5xFAD//Ugcgf/f//Thy1-CreERT2//EYFP mice), when compared to 5xFAD//Ugcgf/f littermates (5xFAD mice). Aberrantly activated glial cells and their expression of pro-inflammatory cytokines have emerged as the major culprits for synaptic loss in AD. Typically, astrocytic activation is accompanied by a thickening of astrocytic processes, which impairs astrocytic support for neuronal synapses. In contrast to 5xFAD mice, 5xFAD//Ugcgf/f//Thy1-CreERT2//EYFP display a less pronounced thickening of astrocytic processes and a lower expression of tumor necrosis factor-α and interleukin 1-α in the hippocampus. Thus, this work further emphasizes that GCS inhibition may constitute a potential therapeutic target against AD.
Collapse
Affiliation(s)
- Silke Herzer
- Division of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Cassidy Hagan
- Division of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany.,Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Johanna von Gerichten
- Division of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany.,Lipid Pathobiochemistry Group, German Cancer Research Center, Heidelberg, Germany
| | - Vanessa Dieterle
- Division of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Bogdan Munteanu
- Center for Mass Spectrometry (CeMOS), University of Heidelberg and Mannheim University of Applied Sciences, Mannheim, Germany
| | - Roger Sandhoff
- Division of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany.,Lipid Pathobiochemistry Group, German Cancer Research Center, Heidelberg, Germany
| | - Carsten Hopf
- Center for Mass Spectrometry (CeMOS), University of Heidelberg and Mannheim University of Applied Sciences, Mannheim, Germany
| | - Viola Nordström
- Division of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
34
|
Hunter S, Smailagic N, Brayne C. Dementia Research: Populations, Progress, Problems, and Predictions. J Alzheimers Dis 2018; 64:S119-S143. [DOI: 10.3233/jad-179927] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sally Hunter
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Nadja Smailagic
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Institute of Public Health, University of Cambridge, Cambridge, UK
| |
Collapse
|
35
|
Cholesterol ester hydrolase inhibitors reduce the production of synaptotoxic amyloid-β oligomers. Biochim Biophys Acta Mol Basis Dis 2018; 1864:649-659. [DOI: 10.1016/j.bbadis.2017.12.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 11/17/2017] [Accepted: 12/11/2017] [Indexed: 11/20/2022]
|
36
|
Rahimi F. Aptamers Selected for Recognizing Amyloid β-Protein-A Case for Cautious Optimism. Int J Mol Sci 2018; 19:ijms19030668. [PMID: 29495486 PMCID: PMC5877529 DOI: 10.3390/ijms19030668] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/18/2018] [Accepted: 02/22/2018] [Indexed: 02/07/2023] Open
Abstract
Aptamers are versatile oligonucleotide ligands used for molecular recognition of diverse targets. However, application of aptamers to the field of amyloid β-protein (Aβ) has been limited so far. Aβ is an intrinsically disordered protein that exists in a dynamic conformational equilibrium, presenting time-dependent ensembles of short-lived, metastable structures and assemblies that have been generally difficult to isolate and characterize. Moreover, despite understanding of potential physiological roles of Aβ, this peptide has been linked to the pathogenesis of Alzheimer disease, and its pathogenic roles remain controversial. Accumulated scientific evidence thus far highlights undesirable or nonspecific interactions between selected aptamers and different Aβ assemblies likely due to the metastable nature of Aβ or inherent affinity of RNA oligonucleotides to β-sheet-rich fibrillar structures of amyloidogenic proteins. Accordingly, lessons drawn from Aβ–aptamer studies emphasize that purity and uniformity of the protein target and rigorous characterization of aptamers’ specificity are important for realizing and garnering the full potential of aptamers selected for recognizing Aβ or other intrinsically disordered proteins. This review summarizes studies of aptamers selected for recognizing different Aβ assemblies and highlights controversies, difficulties, and limitations of such studies.
Collapse
Affiliation(s)
- Farid Rahimi
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia.
| |
Collapse
|
37
|
Duyckaerts C, Sazdovitch V, Ando K, Seilhean D, Privat N, Yilmaz Z, Peckeu L, Amar E, Comoy E, Maceski A, Lehmann S, Brion JP, Brandel JP, Haïk S. Neuropathology of iatrogenic Creutzfeldt-Jakob disease and immunoassay of French cadaver-sourced growth hormone batches suggest possible transmission of tauopathy and long incubation periods for the transmission of Abeta pathology. Acta Neuropathol 2018; 135:201-212. [PMID: 29209767 DOI: 10.1007/s00401-017-1791-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 12/31/2022]
Abstract
Abeta deposits and tau pathology were investigated in 24 French patients that died from iatrogenic Creutzfeldt-Jakob disease after exposure to cadaver-derived human growth hormone (c-hGH) in the 1980s. Abeta deposits were found only in one case that had experienced one of the longest incubation periods. Three cases had also intracellular tau accumulation. The analysis of 24 batches of c-hGH, produced between 1974 and 1988, demonstrated for the first time the presence of Abeta and tau contaminants in c-hGH (in 17 and 6 batches, respectively). The incubation of prion disease was shorter in the French patients than the incubation times reported in two previously published British series. We interpreted the low incidence of Abeta in this French series as a consequence of the shorter incubation period observed in France, as compared to that observed in the United Kingdom. This concept suggested that a mean incubation period for the development of detectable Abeta deposits would be longer than 18 years after the first exposure. Moreover, we hypothesized that tau pathology might also be transmissible in humans.
Collapse
Affiliation(s)
- Charles Duyckaerts
- Inserm U1127, CNRS UMR 7225, UPMC Univ Paris VI UMR S 1127, Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, 47 boulevard de l'Hôpital, 75013, Paris, France.
- Laboratoire de Neuropathologie R. Escourolle, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France.
| | - Véronique Sazdovitch
- Inserm U1127, CNRS UMR 7225, UPMC Univ Paris VI UMR S 1127, Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, 47 boulevard de l'Hôpital, 75013, Paris, France
- Laboratoire de Neuropathologie R. Escourolle, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Kunie Ando
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Danielle Seilhean
- Inserm U1127, CNRS UMR 7225, UPMC Univ Paris VI UMR S 1127, Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, 47 boulevard de l'Hôpital, 75013, Paris, France
- Laboratoire de Neuropathologie R. Escourolle, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Nicolas Privat
- Inserm U1127, CNRS UMR 7225, UPMC Univ Paris VI UMR S 1127, Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, 47 boulevard de l'Hôpital, 75013, Paris, France
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurène Peckeu
- Inserm U1127, CNRS UMR 7225, UPMC Univ Paris VI UMR S 1127, Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, 47 boulevard de l'Hôpital, 75013, Paris, France
| | - Elodie Amar
- Service de Biochimie et Biologie Moléculaire, Hôpital Lariboisière, AP-HP, Paris, France
| | - Emmanuel Comoy
- Commissariat à l'Energie Atomique, DRF/iMETI/SEPIA, Fontenay-aux-Roses, France
| | - Aleksandra Maceski
- Laboratoire de Biochimie Protéomique Clinique, CHU de Montpellier, CRB, INSERM U1183, Université de Montpellier, Montpellier, France
| | - Sylvain Lehmann
- Laboratoire de Biochimie Protéomique Clinique, CHU de Montpellier, CRB, INSERM U1183, Université de Montpellier, Montpellier, France
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, ULB Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Philippe Brandel
- Inserm U1127, CNRS UMR 7225, UPMC Univ Paris VI UMR S 1127, Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, 47 boulevard de l'Hôpital, 75013, Paris, France
- Hôpital de la Pitié-Salpêtrière, Cellule nationale de référence des MCJ, AP-HP, Paris, France
| | - Stéphane Haïk
- Inserm U1127, CNRS UMR 7225, UPMC Univ Paris VI UMR S 1127, Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, 47 boulevard de l'Hôpital, 75013, Paris, France.
- Laboratoire de Neuropathologie R. Escourolle, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France.
- Hôpital de la Pitié-Salpêtrière, Cellule nationale de référence des MCJ, AP-HP, Paris, France.
| |
Collapse
|
38
|
Hunter S, Brayne C. Understanding the roles of mutations in the amyloid precursor protein in Alzheimer disease. Mol Psychiatry 2018; 23:81-93. [PMID: 29112196 DOI: 10.1038/mp.2017.218] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 12/16/2022]
Abstract
Many models of disease progression in Alzheimer's disease (AD) have been proposed to help guide experimental design and aid the interpretation of results. Models focussing on the genetic evidence include the amyloid cascade (ACH) and presenilin (PSH) hypotheses and the amyloid precursor protein (APP) matrix approach (AMA), of which the ACH has held a dominant position for over two decades. However, the ACH has never been fully accepted and has not yet delivered on its therapeutic promise. We review the ACH, PSH and AMA in relation to levels of APP proteolytic fragments reported from AD-associated mutations in APP. Different APP mutations have diverse effects on the levels of APP proteolytic fragments. This evidence is consistent with at least three disease pathways that can differ between familial and sporadic AD and two pathways associated with cerebral amyloid angiopathy. We cannot fully evaluate the ACH, PSH and AMA in relation to the effects of mutations in APP as the APP proteolytic system has not been investigated systematically. The confounding effects of sequence homology, complexity of competing cleavages and antibody cross reactivities all illustrate limitations in our understanding of the roles these fragments and the APP proteolytic system as a whole in normal aging and disease play. Current experimental design should be refined to generate clearer evidence, addressing both aging and complex disorders with standardised reporting formats. A more flexible theoretical framework capable of accommodating the complexity of the APP proteolytic system is required to integrate available evidence.
Collapse
Affiliation(s)
- S Hunter
- Department of Public Health and Primary Care, Institute of Public Health, Forvie Site University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - C Brayne
- Department of Public Health and Primary Care, Institute of Public Health, Forvie Site University of Cambridge, School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
39
|
Bate C, Williams A. Monomeric amyloid-β reduced amyloid-β oligomer-induced synapse damage in neuronal cultures. Neurobiol Dis 2017; 111:48-58. [PMID: 29272738 DOI: 10.1016/j.nbd.2017.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 11/12/2017] [Accepted: 12/12/2017] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disease characterized by the accumulation of amyloid-β (Aβ) in the brain. Aβ oligomers are believed to cause synapse damage resulting in the memory deficits that are characteristic of this disease. Since the loss of synaptic proteins in the brain correlates closely with the degree of dementia in Alzheimer's disease, the process of Aβ-induced synapse damage was investigated in cultured neurons by measuring the loss of synaptic proteins. Soluble Aβ oligomers, derived from Alzheimer's-affected brains, caused the loss of cysteine string protein and synaptophysin from neurons. When applied to synaptosomes Aβ oligomers increased cholesterol concentrations and caused aberrant activation of cytoplasmic phospholipase A2 (cPLA2). In contrast, Aβ monomer preparations did not affect cholesterol concentrations or activate synaptic cPLA2, nor did they damage synapses. The Aβ oligomer-induced aggregation of cellular prion proteins (PrPC) at synapses triggered the activation of cPLA2 that leads to synapse degeneration. Critically, Aβ monomer preparations did not cause the aggregation of PrPC; rather they reduced the Aβ oligomer-induced aggregation of PrPC. The presence of Aβ monomer preparations also inhibited the Aβ oligomer-induced increase in cholesterol concentrations and activation of cPLA2 in synaptosomes and protected neurons against the Aβ oligomer-induced synapse damage. These results support the hypothesis that Aβ monomers are neuroprotective. We hypothesise that synapse damage may result from a pathological Aβ monomer:oligomer ratio rather than the total concentrations of Aβ within the brain.
Collapse
Affiliation(s)
- Clive Bate
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Herts AL9 7TA, UK.
| | - Alun Williams
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK.
| |
Collapse
|
40
|
Hunter S, Brayne C. Erratum to: Do anti-amyloid beta protein antibody cross reactivities confound Alzheimer disease research? J Negat Results Biomed 2017; 16:8. [PMID: 28274242 PMCID: PMC5343297 DOI: 10.1186/s12952-017-0073-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Sally Hunter
- Department of Public Health and Primary Care, Institute of Public Health Forvie Site, University of Cambridge School of Clinical Medicine, Box 113 Cambridge Biomedical Campus, Cambridge, CB2 0SP, UK.
| | - Carol Brayne
- Department of Public Health and Primary Care, Institute of Public Health Forvie Site, University of Cambridge School of Clinical Medicine, Box 113 Cambridge Biomedical Campus, Cambridge, CB2 0SP, UK
| |
Collapse
|