1
|
Kuznetsov AV. Evaluating the Combined Neurotoxicity of Amyloid Beta and Tau Oligomers in Alzheimer's Disease: A Novel Cellular-Level Criterion. J Biomech Eng 2025; 147:041003. [PMID: 39840747 DOI: 10.1115/1.4067701] [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: 11/03/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
A criterion characterizing the combined neurotoxicity of amyloid beta and tau oligomers is suggested. A mathematical model for calculating the value of this criterion during senile plaque and neurofibrillary tangle (NFT) formation is proposed. Computations show that for physiologically relevant parameter values, the value of the criterion increases approximately linearly with time. Once neurofibrillary tangles begin forming in addition to senile plaques, there is an increase in the slope characterizing the rate at which the criterion increases. The critical value of the criterion at which a neuron dies is estimated. Unless the production rates of amyloid beta and tau monomers are very large, computations predict that for the accumulated toxicity to reach the critical value, the neural machinery responsible for the degradation of amyloid beta and tau monomers and aggregates must become dysfunctional. The value of the criterion after 20 years of the aggregation process is strongly influenced by the deposition rates of amyloid beta and tau oligomers into senile plaques and NFTs. This suggests that deposition of amyloid beta and tau oligomers into senile plaques and NFTs may reduce accumulated toxicity by sequestering more toxic oligomeric species into less toxic insoluble aggregates.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910
| |
Collapse
|
2
|
Kuznetsov AV. Effect of diffusivity of amyloid beta monomers on the formation of senile plaques. MATHEMATICAL MEDICINE AND BIOLOGY : A JOURNAL OF THE IMA 2024; 41:346-362. [PMID: 39404062 DOI: 10.1093/imammb/dqae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 12/17/2024]
Abstract
Alzheimer's disease (AD) presents a perplexing question: why does its development span decades, even though individual amyloid beta (Aβ) deposits (senile plaques) can form rapidly in as little as 24 hours, as recent publications suggest? This study investigated whether the formation of senile plaques can be limited by factors other than polymerization kinetics alone. Instead, their formation may be limited by the diffusion-driven supply of Aβ monomers, along with the rate at which the monomers are produced from amyloid precursor protein and the rate at which Aβ monomers undergo degradation. A mathematical model incorporating the nucleation and autocatalytic process (via the Finke-Watzky model), as well as Aβ monomer diffusion, was proposed. The obtained system of partial differential equations was solved numerically, and a simplified version was investigated analytically. The computational results predicted that it takes approximately 7 years for Aβ aggregates to reach a neurotoxic concentration of 50 μM. Additionally, a sensitivity analysis was performed to examine how the diffusivity of Aβ monomers and their production rate impact the concentration of Aβ aggregates.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, 1840 Entrepreneur Drive, Raleigh, North Carolina 27695-7910, USA
| |
Collapse
|
3
|
Hossen F, Geng X, Sun GY, Yao X, Lee JC. Oligomeric Amyloid-β and Tau Alter Cell Adhesion Properties and Induce Inflammatory Responses in Cerebral Endothelial Cells Through the RhoA/ROCK Pathway. Mol Neurobiol 2024; 61:8759-8776. [PMID: 38561558 PMCID: PMC11445398 DOI: 10.1007/s12035-024-04138-z] [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: 07/03/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
Dysfunction of cerebral endothelial cells (CECs) has been implicated in the pathology of Alzheimer's disease (AD). Despite evidence showing cytotoxic effects of oligomeric amyloid-β (oAβ) and Tau (oTau) in the central nervous system, their direct effects on CECs have not been fully investigated. In this study, we examined the direct effects of oAβ, oTau, and their combination on cell adhesion properties and inflammatory responses in CECs. We found that both oAβ and oTau increased cell stiffness, as well as the p-selectin/Sialyl-LewisX (sLeX) bonding-mediated membrane tether force and probability of adhesion in CECs. Consistent with these biomechanical alterations, treatments with oAβ or oTau also increased actin polymerization and the expression of p-selectin at the cell surface. These toxic oligomeric peptides also triggered inflammatory responses, including upregulations of p-NF-kB p65, IL-1β, and TNF-α. In addition, they rapidly activated the RhoA/ROCK pathway. These biochemical and biomechanical changes were further enhanced by the treatment with the combination of oAβ and oTau, which were significantly suppressed by Fasudil, a specific inhibitor for the RhoA/ROCK pathway. In conclusion, our data suggest that oAβ, oTau, and their combination triggered subcellular mechanical alterations and inflammatory responses in CECs through the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Faruk Hossen
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Xue Geng
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Xincheng Yao
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - James C Lee
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
4
|
Kuznetsov AV. The growth rate of senile plaques is determined by the competition between the rate of deposition of free Aβ aggregates into plaques and the autocatalytic production of free Aβ aggregates. J Theor Biol 2024; 593:111900. [PMID: 38992461 DOI: 10.1016/j.jtbi.2024.111900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
The formation of amyloid beta (Aβ) deposits (senile plaques) is one of the hallmarks of Alzheimer's disease (AD). This study investigates what processes are primarily responsible for their formation. A model is developed to simulate the diffusion of amyloid beta (Aβ) monomers, the production of free Aβ aggregates through nucleation and autocatalytic processes, and the deposition of these aggregates into senile plaques. The model suggests that efficient degradation of Aβ monomers alone may suffice to prevent the growth of senile plaques, even without degrading Aβ aggregates and existing plaques. This is because the degradation of Aβ monomers interrupts the supply of reactants needed for plaque formation. The impact of Aβ monomer diffusivity is demonstrated to be small, enabling the application of the lumped capacitance approximation and the derivation of approximate analytical solutions for limiting cases with both small and large rates of Aβ aggregate deposition into plaques. It is found that the rate of plaque growth is governed by two competing processes. One is the deposition rate of free Aβ aggregates into senile plaques. If this rate is small, the plaque grows slowly. However, if the rate of deposition of Aβ aggregates into senile plaques is very large, the free Aβ aggregates are removed from the intracellular fluid by deposition into the plaques, leaving insufficient free Aβ aggregates to catalyze the production of new aggregates. This suggests that under certain conditions, Aβ plaques may offer neuroprotection and impede their own growth. Additionally, it indicates that there exists an optimal rate of deposition of free Aβ aggregates into the plaques, at which the plaques attain their maximum size.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA.
| |
Collapse
|
5
|
Kuznetsov AV. Numerical modeling of senile plaque development under conditions of limited diffusivity of amyloid-β monomers. J Theor Biol 2024; 587:111823. [PMID: 38608804 DOI: 10.1016/j.jtbi.2024.111823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024]
Abstract
This paper introduces a new model to simulate the progression of senile plaques, focusing on scenarios where concentrations of amyloid beta (Aβ) monomers and aggregates vary between neurons. Extracellular variations in these concentrations may arise due to limited diffusivity of Aβ monomers and a high rate of Aβ monomer production at lipid membranes, requiring a substantial concentration gradient for diffusion-driven transport of Aβ monomers. The dimensionless formulation of the model is presented, which identifies four key dimensionless parameters governing the solutions for Aβ monomer and aggregate concentrations, as well as the radius of a growing Aβ plaque within the control volume. These parameters include the dimensionless diffusivity of Aβ monomers, the dimensionless rate of Aβ monomer production, and the dimensionless half-lives of Aβ monomers and aggregates. A dimensionless parameter is then introduced to evaluate the validity of the lumped capacitance approximation. An approximate solution is derived for the scenario involving large diffusivity of Aβ monomers and dysfunctional protein degradation machinery, resulting in infinitely long half-lives for Aβ monomers and aggregates. In this scenario, the concentrations of Aβ aggregates and the radius of the Aβ plaque depend solely on a single dimensionless parameter that characterizes the rate of Aβ monomer production. According to the approximate solution, the concentration of Aβ aggregates is linearly dependent on the rate of monomer production, and the radius of an Aβ plaque is directly proportional to the cube root of the rate of monomer production. However, when departing from the conditions of the approximate solution (e.g., finite half-lives), the concentrations of Aβ monomers and aggregates, along with the plaque radius, exhibit complex dependencies on all four dimensionless parameters. For instance, under physiological half-life conditions, the plaque radius reaches a maximum value and stabilizes thereafter.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA.
| |
Collapse
|
6
|
Kuznetsov AV. Numerical and Analytical Simulation of the Growth of Amyloid-β Plaques. J Biomech Eng 2024; 146:061004. [PMID: 38421364 DOI: 10.1115/1.4064969] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
Numerical and analytical solutions were employed to calculate the radius of an amyloid-β (Aβ) plaque over time. To the author's knowledge, this study presents the first model simulating the growth of Aβ plaques. Findings indicate that the plaque can attain a diameter of 50 μm after 20 years of growth, provided the Aβ monomer degradation machinery is malfunctioning. A mathematical model incorporates nucleation and autocatalytic growth processes using the Finke-Watzky model. The resulting system of ordinary differential equations was solved numerically, and for the simplified case of infinitely long Aβ monomer half-life, an analytical solution was found. Assuming that Aβ aggregates stick together and using the distance between the plaques as an input parameter of the model, it was possible to calculate the plaque radius from the concentration of Aβ aggregates. This led to the "cube root hypothesis," positing that Aβ plaque size increases proportionally to the cube root of time. This hypothesis helps explain why larger plaques grow more slowly. Furthermore, the obtained results suggest that the plaque size is independent of the kinetic constants governing Aβ plaque agglomeration, indicating that the kinetics of Aβ plaque agglomeration is not a limiting factor for plaque growth. Instead, the plaque growth rate is limited by the rates of Aβ monomer production and degradation.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910
| |
Collapse
|
7
|
Cook I, Leyh TS. Sterol-activated amyloid beta fibril formation. J Biol Chem 2023; 299:105445. [PMID: 37949224 PMCID: PMC10704437 DOI: 10.1016/j.jbc.2023.105445] [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: 08/27/2023] [Revised: 10/23/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023] Open
Abstract
The metabolic processes that link Alzheimer's disease (AD) to elevated cholesterol levels in the brain are not fully defined. Amyloid beta (Aβ) plaque accumulation is believed to begin decades prior to symptoms and to contribute significantly to the disease. Cholesterol and its metabolites accelerate plaque formation through as-yet-undefined mechanisms. Here, the mechanism of cholesterol (CH) and cholesterol 3-sulfate (CS) induced acceleration of Aβ42 fibril formation is examined in quantitative ligand binding, Aβ42 fibril polymerization, and molecular dynamics studies. Equilibrium and pre-steady-state binding studies reveal that monomeric Aβ42•ligand complexes form and dissociate rapidly relative to oligomerization, that the ligand/peptide stoichiometry is 1-to-1, and that the peptide is likely saturated in vivo. Analysis of Aβ42 polymerization progress curves demonstrates that ligands accelerate polymer synthesis by catalyzing the conversion of peptide monomers into dimers that nucleate the polymerization reaction. Nucleation is accelerated ∼49-fold by CH, and ∼13,000-fold by CS - a minor CH metabolite. Polymerization kinetic models predict that at presumed disease-relevant CS and CH concentrations, approximately half of the polymerization nuclei will contain CS, small oligomers of neurotoxic dimensions (∼12-mers) will contain substantial CS, and fibril-formation lag times will decrease 13-fold relative to unliganded Aβ42. Molecular dynamics models, which quantitatively predict all experimental findings, indicate that the acceleration mechanism is rooted in ligand-induced stabilization of the peptide in non-helical conformations that readily form polymerization nuclei.
Collapse
Affiliation(s)
- Ian Cook
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Thomas S Leyh
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
8
|
Żyła A, Martel A, Jurczak P, Moliński A, Szymańska A, Kozak M. Human cystatin C induces the disaggregation process of selected amyloid beta peptides: a structural and kinetic view. Sci Rep 2023; 13:20833. [PMID: 38012338 PMCID: PMC10682421 DOI: 10.1038/s41598-023-47514-w] [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: 09/20/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD) and various types of amyloidosis, are incurable; therefore, understanding the mechanisms of amyloid decomposition is crucial to develop an effective drug against them for future therapies. It has been reported that one out of three people over the age of 85 are suffering from dementia as a comorbidity to AD. Amyloid beta (Aβ), the hallmark of AD, transforms structurally from monomers into β-stranded aggregates (fibrils) via multiple oligomeric states. Astrocytes in the central nervous system secrete the human cystatin C protein (HCC) in response to various proteases and cytokines. The codeposition of Aβ and HCC in the brains of patients with AD led to the hypothesis that cystatin C is implicated in the disease process. In this study, we investigate the intermolecular interactions between different atomic structures of fibrils formed by Aβ peptides and HCC to understand the pathological aggregation of these polypeptides into neurotoxic oligomers and then amyloid plaques. To characterize the interactions between Aβ and HCC, we used a complementary approach based on the combination of small-angle neutron scattering analysis, atomic force microscopy and computational modelling, allowing the exploration of the structures of multicomponent protein complexes. We report here an optimized protocol to study that interaction. The results show a dependency of the sequence length of the Aβ peptide on the ability of the associated HCC to disaggregate it.
Collapse
Affiliation(s)
- Adriana Żyła
- Department of Biomedical Physics, Faculty of Physics, Adam Mickiewicz University, Poznań, Poland
- NanoBioMedical Centre, Adam Mickiewicz University, Poznań, Poland
| | - Anne Martel
- Large Scale Structures, ILL Neutrons for Society, Institute Laue-Langevin, Grenoble, France
| | - Przemysław Jurczak
- Laboratory of Medical Chemistry, Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Augustyn Moliński
- Department of Biomedical Physics, Faculty of Physics, Adam Mickiewicz University, Poznań, Poland
- NanoBioMedical Centre, Adam Mickiewicz University, Poznań, Poland
| | - Aneta Szymańska
- Laboratory of Medical Chemistry, Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Maciej Kozak
- Department of Biomedical Physics, Faculty of Physics, Adam Mickiewicz University, Poznań, Poland.
- SOLARIS National Synchrotron Radiation Centre, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
9
|
Rey JA, Farid UM, Najjoum CM, Brown A, Magdoom KN, Mareci TH, Sarntinoranont M. Perivascular network segmentations derived from high-field MRI and their implications for perivascular and parenchymal mass transport in the rat brain. Sci Rep 2023; 13:9205. [PMID: 37280246 DOI: 10.1038/s41598-023-34850-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 05/09/2023] [Indexed: 06/08/2023] Open
Abstract
A custom segmentation workflow was applied to ex vivo high-field MR images of rat brains acquired following in vivo intraventricular contrast agent infusion to generate maps of the perivascular spaces (PVS). The resulting perivascular network segmentations enabled analysis of perivascular connections to the ventricles, parenchymal solute clearance, and dispersive solute transport within PVS. Numerous perivascular connections between the brain surface and the ventricles suggest the ventricles integrate into a PVS-mediated clearance system and raise the possibility of cerebrospinal fluid (CSF) return from the subarachnoid space to the ventricles via PVS. Assuming rapid solute exchange between the PVS and CSF spaces primarily by advection, the extensive perivascular network decreased the mean clearance distance from parenchyma to the nearest CSF compartment resulting in an over 21-fold reduction in the estimated diffusive clearance time scale, irrespective of solute diffusivity. This corresponds to an estimated diffusive clearance time scale under 10 min for amyloid-beta which suggests that the widespread distribution of PVS may render diffusion an effective parenchymal clearance mechanism. Additional analysis of oscillatory solute dispersion within PVS indicates that advection rather than dispersion is likely the primary transport mechanism for dissolved compounds greater than 66 kDa in the long (> 2 mm) perivascular segments identified here, although dispersion may be significant for smaller compounds in shorter perivascular segments.
Collapse
Affiliation(s)
- Julian A Rey
- Department of Mechanical and Aerospace Engineering, University of Florida, PO BOX 116250, Gainesville, FL, 32611, USA
| | - Uzair M Farid
- Department of Mechanical and Aerospace Engineering, University of Florida, PO BOX 116250, Gainesville, FL, 32611, USA
| | - Christopher M Najjoum
- Department of Mechanical and Aerospace Engineering, University of Florida, PO BOX 116250, Gainesville, FL, 32611, USA
| | - Alec Brown
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Kulam Najmudeen Magdoom
- Department of Mechanical and Aerospace Engineering, University of Florida, PO BOX 116250, Gainesville, FL, 32611, USA
| | - Thomas H Mareci
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Malisa Sarntinoranont
- Department of Mechanical and Aerospace Engineering, University of Florida, PO BOX 116250, Gainesville, FL, 32611, USA.
| |
Collapse
|
10
|
Wenzel TJ, Le J, He J, Alcorn J, Mousseau DD. Fundamental Neurochemistry Review: Incorporating a greater diversity of cell types, including microglia, in brain organoid cultures improves clinical translation. J Neurochem 2023; 164:560-582. [PMID: 36517959 DOI: 10.1111/jnc.15741] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Brain organoids have the potential to improve clinical translation, with the added benefit of reducing any extraneous use of experimental animals. As brain organoids are three-dimensional in vitro constructs that emulate the human brain, they bridge in vitro and in vivo studies more appropriately than monocultures. Although many factors contribute to the failure of extrapolating monoculture-based information to animal-based experiments and clinical trials, for the purpose of this review, we will focus on glia (non-neuronal brain cells), whose functions and transcriptome are particularly abnormal in monocultures. As discussed herein, glia require signals from-and contact with-other cell types to exist in their homeostatic state, which likely contributes to some of the differences between data derived from monocultures and data derived from brain organoids and even two-dimensional co-cultures. Furthermore, we highlight transcriptomic differences between humans and mice in regard to aging and Alzheimer's disease, emphasizing need for a model using the human genome-again, a benefit of brain organoids-to complement data derived from animals. We also identify an urgency for guidelines to improve the reporting and transparency of research using organoids. The lack of reporting standards creates challenges for the comparison and discussion of data from different articles. Importantly, brain organoids mark the first human model enabling the study of brain cytoarchitecture and development.
Collapse
Affiliation(s)
- Tyler J Wenzel
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jennifer Le
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jim He
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jane Alcorn
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
11
|
Wenzel TJ, Murray TE, Noyovitz B, Narayana K, Gray TE, Le J, He J, Simtchouk S, Gibon J, Alcorn J, Mousseau DD, Zandberg WF, Klegeris A. Cardiolipin released by microglia can act on neighboring glial cells to facilitate the uptake of amyloid-β (1-42). Mol Cell Neurosci 2023; 124:103804. [PMID: 36592800 DOI: 10.1016/j.mcn.2022.103804] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/16/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
Cardiolipin is a mitochondrial phospholipid that is also detected in serum inferring its extracellular release; however, this process has not been directly demonstrated for any of the brain cell types. Nevertheless, extracellular cardiolipin has been shown to modulate several neuroimmune functions of microglia and astrocytes, including upregulation of their endocytic activity. Low cardiolipin levels are associated with brain aging, and may thus hinder uptake of amyloid-β (Αβ) in Alzheimer's disease. We hypothesized that glial cells are one of the sources of extracellular cardiolipin in the brain parenchyma where this phospholipid interacts with neighboring cells to upregulate the endocytosis of Αβ. Liquid chromatography-mass spectrophotometry identified 31 different species of cardiolipin released from murine BV-2 microglial cells and revealed this process was accelerated by exposure to Aβ42. Extracellular cardiolipin upregulated internalization of fluorescently-labeled Aβ42 by primary murine astrocytes, human U118 MG astrocytic cells, and murine BV-2 microglia. Increased endocytic activity in the presence of extracellular cardiolipin was also demonstrated by studying uptake of Aβ42 and pHrodo™ Bioparticles™ by human induced pluripotent stem cells (iPSCs)-derived microglia, as well as iPSC-derived human brain organoids containing microglia, astrocytes, oligodendrocytes and neurons. Our observations indicate that Aβ42 augments the release of cardiolipin from microglia into the extracellular space, where it can act on microglia and astrocytes to enhance their endocytosis of Aβ42. Our observations suggest that the reduced glial uptake of Aβ due to the decreased levels of cardiolipin could be at least partially responsible for the extracellular accumulation of Aβ in aging and Alzheimer's disease.
Collapse
Affiliation(s)
- Tyler J Wenzel
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada; College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Taryn E Murray
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Benjamin Noyovitz
- Department of Chemistry, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Kamal Narayana
- Department of Chemistry, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Taylor E Gray
- Department of Chemistry, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Jennifer Le
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - Jim He
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - Svetlana Simtchouk
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Julien Gibon
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Jane Alcorn
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| | - Wesley F Zandberg
- Department of Chemistry, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada.
| |
Collapse
|
12
|
Pönisch W, Michaels TCT, Weber CA. Aggregation controlled by condensate rheology. Biophys J 2023; 122:197-214. [PMID: 36369755 PMCID: PMC9822804 DOI: 10.1016/j.bpj.2022.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 06/12/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022] Open
Abstract
Biomolecular condensates in living cells can exhibit a complex rheology, including viscoelastic and glassy behavior. This rheological behavior of condensates was suggested to regulate polymerization of cytoskeletal filaments and aggregation of amyloid fibrils. Here, we theoretically investigate how the rheological properties of condensates can control the formation of linear aggregates. To this end, we propose a kinetic theory for linear aggregation in coexisting phases, which accounts for the aggregate size distribution and the exchange of aggregates between inside and outside of condensates. The rheology of condensates is accounted in our model via aggregate mobilities that depend on aggregate size. We show that condensate rheology determines whether aggregates of all sizes or dominantly small aggregates are exchanged between condensate inside and outside on the timescale of aggregation. As a result, the ratio of aggregate numbers inside to outside of condensates differs significantly. Strikingly, we also find that weak variations in the rheological properties of condensates can lead to a switch-like change of the number of aggregates. These results suggest a possible physical mechanism for how living cells could control linear aggregation in a switch-like fashion through variations in condensate rheology.
Collapse
Affiliation(s)
- Wolfram Pönisch
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Thomas C T Michaels
- Laboratory for Molecular Cell Biology, University College London, London, United Kingdom; Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, United Kingdom; Department of Biology, Institute of Biochemistry, ETH Zurich, Zurich, Switzerland.
| | - Christoph A Weber
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany; Faculty of Mathematics, Natural Sciences, and Materials Engineering, Institute of Physics, University of Augsburg, Augsburg, Germany.
| |
Collapse
|
13
|
Yavorsky VA, Rozumna NM, Lukyanetz EA. Influence of amyloid beta on impulse spiking of isolated hippocampal neurons. Front Cell Neurosci 2023; 17:1132092. [PMID: 37124394 PMCID: PMC10133472 DOI: 10.3389/fncel.2023.1132092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
One of the signs of Alzheimer's disease (AD) is the formation of β-amyloid plaques, which ultimately lead to the dysfunction of neurons with subsequent neurodegeneration. Although extensive researches have been conducted on the effects of different amyloid conformations such as oligomers and fibrils on neuronal function in isolated cells and circuits, the exact contribution of extracellular beta-amyloid on neurons remains incompletely comprehended. In our experiments, we studied the effect of β-amyloid peptide (Aβ1-42) on the action potential (APs) generation in isolated CA1 hippocampal neurons in perforated patch clamp conditions. Our findings demonstrate that Aβ1-42 affects the generation of APs differently in various hippocampal neurons, albeit with a shared effect of enhancing the firing response of the neurons within a minute of the start of Aβ1-42 application. In the first response type, there was a shift of 20-65% toward smaller values in the firing threshold of action potentials in response to inward current. Conversely, the firing threshold of action potentials was not affected in the second type of response to the application of Aβ1-42. In these neurons, Aβ1-42 caused a moderate increase in the frequency of spiking, up to 15%, with a relatively uniform increase in the frequency of action potentials generation regardless of the level of input current. Obtained data prove the absence of direct short-term negative effect of the Aβ1-42 on APs generation in neurons. Even with increasing the APs generation frequency and lowering the neurons' activation threshold, neurons were functional. Obtained data can suggest that only the long-acting presence of the Aβ1-42 in the cell environment can cause neuronal dysfunction due to a prolonged increase of APs firing and predisposition to this process.
Collapse
|
14
|
Lee WJ, Brown JA, Kim HR, La Joie R, Cho H, Lyoo CH, Rabinovici GD, Seong JK, Seeley WW. Regional Aβ-tau interactions promote onset and acceleration of Alzheimer's disease tau spreading. Neuron 2022; 110:1932-1943.e5. [PMID: 35443153 PMCID: PMC9233123 DOI: 10.1016/j.neuron.2022.03.034] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/19/2022] [Accepted: 03/28/2022] [Indexed: 12/22/2022]
Abstract
Amyloid-beta and tau are key molecules in the pathogenesis of Alzheimer's disease, but it remains unclear how these proteins interact to promote disease. Here, by combining cross-sectional and longitudinal molecular imaging and network connectivity analyses in living humans, we identified two amyloid-beta/tau interactions associated with the onset and propagation of tau spreading. First, we show that the lateral entorhinal cortex, an early site of tau neurofibrillary tangle formation, is subject to remote, connectivity-mediated amyloid-beta/tau interactions linked to initial tau spreading. Second, we identify the inferior temporal gyrus as the region featuring the greatest local amyloid-beta/tau interactions and a connectivity profile well suited to accelerate tau propagation. Taken together, our data address long-standing questions regarding the topographical dissimilarity between early amyloid-beta and tau deposition.
Collapse
Affiliation(s)
- Wha Jin Lee
- School of Biomedical Engineering, Korea University, Seoul 02841, South Korea
| | - Jesse A Brown
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hye Ryun Kim
- School of Biomedical Engineering, Korea University, Seoul 02841, South Korea; Global Health Technology Research Center, College of Health Science, Korea University, Seoul 02841, South Korea
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Seoul 06273, South Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Seoul 06273, South Korea
| | - Gil D Rabinovici
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joon-Kyung Seong
- School of Biomedical Engineering, Korea University, Seoul 02841, South Korea; Department of Artificial Intelligence, Korea University, Seoul 02841, South Korea.
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
15
|
Coupled Neural–Glial Dynamics and the Role of Astrocytes in Alzheimer’s Disease. MATHEMATICAL AND COMPUTATIONAL APPLICATIONS 2022. [DOI: 10.3390/mca27030033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Neurodegenerative diseases such as Alzheimer’s (AD) are associated with the propagation and aggregation of toxic proteins. In the case of AD, it was Alzheimer himself who showed the importance of both amyloid beta (Aβ) plaques and tau protein neurofibrillary tangles (NFTs) in what he called the “disease of forgetfulness”. The amyloid beta forms extracellular aggregates and plaques, whereas tau proteins are intracellular proteins that stabilize axons by cross-linking microtubules that can form largely messy tangles. On the other hand, astrocytes and microglial cells constantly clear these plaques and NFTs from the brain. Astrocytes transport nutrients from the blood to neurons. Activated astrocytes produce monocyte chemoattractant protein-1 (MCP-1), which attracts anti-inflammatory macrophages and clears Aβ. At the same time, the microglia cells are poorly phagocytic for Aβ compared to proinflammatory and anti-inflammatory macrophages. In addition to such distinctive neuropathological features of AD as amyloid beta and tau proteins, neuroinflammation has to be brought into the picture as well. Taking advantage of a coupled mathematical modelling framework, we formulate a network model, accounting for the coupling between neurons and astroglia and integrating all three main neuropathological features with the brain connectome data. We provide details on the coupled dynamics involving cytokines, astrocytes, and microglia. Further, we apply the tumour necrosis factor alpha (TNF-α) inhibitor and anti-Aβ drug and analyze their influence on the brain cells, suggesting conditions under which the drug can prevent cell damage. The important role of astrocytes and TNF-α inhibitors in AD pathophysiology is emphasized, along with potentially promising pathways for developing new AD therapies.
Collapse
|
16
|
Timakwe S, Silwana B, Matoetoe MC. Electrochemistry as a Complementary Technique for Revealing the Influence of Reducing Agent Concentration on AgNPs. ACS OMEGA 2022; 7:4921-4931. [PMID: 35187311 PMCID: PMC8851659 DOI: 10.1021/acsomega.1c05374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/23/2021] [Indexed: 05/23/2023]
Abstract
The synthesis process of AgNPs has been attracting a lot of attention in the fields of biosensors/sensors, diagnostics, and therapeutic applications. An attempt to understand the effect of different concentrations of reducing agents on the synthetic design process has been made. In this paper, we gather information on voltammetry studies and relate it with UV-vis and scanning electron microscopy (SEM) analyses. Given the kinetics, localized surface plasmon absorption (LSPR) band, and narrow size distribution of these methods, it was possible to compare the obtained measurements and clearly distinguish sizes and aggregation. AgNPs measured by SEM showed a statistically significant reduction of the nanoparticle sizes from 65 to 37.5 nm as the reducing agent increased. Well-matched d-spacing data calculated from selected area electron diffraction (SAED) patterns and X-ray diffraction (XRD) were obtained for all of the samples. The UV-vis studies showed that the SPR bands shift toward the blue region as the reducing agent concentration is increased, indicating a decrease in particle sizes. It is worth emphasizing that cyclic voltammetry (CV) and differential pulse voltammetry (DPV) coincide well with SEM on the aggregation of AgNPs at higher concentrations. A 10 mM reducing agent concentration resulted in uniform outcomes for producing AgNPs with the smallest size in terms of full width at half-maximum (FWHM) in all of the methods used in this study, while UV-vis band gaps increase with increasing reducing agent concentration. In agreement with all of the methods investigated, the results suggested that the best concentration of the reducing agents is 10 mM for a target application. These findings suggest the usefulness of voltammetry as a complementary method that can be used as a qualitative guide to identify the size and aggregation of NPs.
Collapse
|
17
|
Cheng KC, Chen YH, Wu CL, Lee WP, Cheung CHA, Chiang HC. Rac1 and Akt Exhibit Distinct Roles in Mediating Aβ-Induced Memory Damage and Learning Impairment. Mol Neurobiol 2021; 58:5224-5238. [PMID: 34273104 DOI: 10.1007/s12035-021-02471-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/24/2021] [Indexed: 11/25/2022]
Abstract
Accumulated beta-amyloid (Aβ) in the brain is the hallmark of Alzheimer's disease (AD). Despite Aβ accumulation is known to trigger cellular dysfunctions and learning and memory damage, the detailed molecular mechanism remains elusive. Recent studies have shown that the onset of memory impairment and learning damage in the AD animal is different, suggesting that the underlying mechanism of the development of memory impairment and learning damage may not be the same. In the current study, with the use of Aβ42 transgenic flies as models, we found that Aβ induces memory damage and learning impairment via differential molecular signaling pathways. In early stage, Aβ activates both Ras and PI3K to regulate Rac1 activity, which affects mostly on memory performance. In later stage, PI3K-Akt is strongly activated by Aβ, which leads to learning damage. Moreover, reduced Akt, but not Rac1, activity promotes cell viability in the Aβ42 transgenic flies, indicating that Akt and Rac1 exhibit differential roles in Aβ regulating toxicity. Taken together, different molecular and cellular mechanisms are involved in Aβ-induced learning damage and memory decline; thus, caution should be taken during the development of therapeutic intervention in the future.
Collapse
Affiliation(s)
- Kuan-Chung Cheng
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
| | - Ying-Hao Chen
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Lin Wu
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Wang-Pao Lee
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun Hei Antonio Cheung
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
| | - Hsueh-Cheng Chiang
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan, Taiwan.
| |
Collapse
|
18
|
McFarland KN, Ceballos C, Rosario A, Ladd T, Moore B, Golde G, Wang X, Allen M, Ertekin-Taner N, Funk CC, Robinson M, Baloni P, Rappaport N, Chakrabarty P, Golde TE. Microglia show differential transcriptomic response to Aβ peptide aggregates ex vivo and in vivo. Life Sci Alliance 2021; 4:4/7/e202101108. [PMID: 34127518 PMCID: PMC8321667 DOI: 10.26508/lsa.202101108] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 01/01/2023] Open
Abstract
Aggregation and accumulation of amyloid-β (Aβ) is a defining feature of Alzheimer's disease pathology. To study microglial responses to Aβ, we applied exogenous Aβ peptide, in either oligomeric or fibrillar conformation, to primary mouse microglial cultures and evaluated system-level transcriptional changes and then compared these with transcriptomic changes in the brains of CRND8 APP mice. We find that primary microglial cultures have rapid and massive transcriptional change in response to Aβ. Transcriptomic responses to oligomeric or fibrillar Aβ in primary microglia, although partially overlapping, are distinct and are not recapitulated in vivo where Aβ progressively accumulates. Furthermore, although classic immune mediators show massive transcriptional changes in the primary microglial cultures, these changes are not observed in the mouse model. Together, these data extend previous studies which demonstrate that microglia responses ex vivo are poor proxies for in vivo responses. Finally, these data demonstrate the potential utility of using microglia as biosensors of different aggregate conformation, as the transcriptional responses to oligomeric and fibrillar Aβ can be distinguished.
Collapse
Affiliation(s)
- Karen N McFarland
- Department of Neurology, University of Florida, Gainesville, FL, USA .,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Carolina Ceballos
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Awilda Rosario
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Thomas Ladd
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Brenda Moore
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Griffin Golde
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Xue Wang
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.,Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Cory C Funk
- Institute for Systems Biology, Seattle, WA, USA
| | | | | | | | - Paramita Chakrabarty
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Todd E Golde
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA .,McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, USA
| |
Collapse
|
19
|
Simpson L, Szeto GL, Boukari H, Good TA, Leach JB. Impact of Four Common Hydrogels on Amyloid-β (Aβ) Aggregation and Cytotoxicity: Implications for 3D Models of Alzheimer's Disease. ACS OMEGA 2020; 5:20250-20260. [PMID: 32832778 PMCID: PMC7439392 DOI: 10.1021/acsomega.0c02046] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/23/2020] [Indexed: 05/08/2023]
Abstract
The physiochemical properties of hydrogels utilized in 3D culture can be used to modulate cell phenotype and morphology with a striking resemblance to cellular processes that occur in vivo. Indeed, research areas including regenerative medicine, tissue engineering, in vitro cancer models, and stem cell differentiation have readily utilized 3D biomaterials to investigate cell biological questions. However, cells are only one component of this biomimetic milieu. In many models of disease such as Alzheimer's disease (AD) that could benefit from the in vivo-like cell morphology associated with 3D culture, other aspects of the disease such as protein aggregation have yet to be methodically considered in this 3D context. A hallmark of AD is the accumulation of the peptide amyloid-β (Aβ), whose aggregation is associated with neurotoxicity. We have previously demonstrated the attenuation of Aβ cytotoxicity when cells were cultured within type I collagen hydrogels versus on 2D substrates. In this work, we investigated the extent to which this phenomenon is conserved when Aβ is confined within hydrogels of varying physiochemical properties, notably mesh size and bioactivity. We investigated the Aβ structure and aggregation kinetics in solution and hydrogels composed of type I collagen, agarose, hyaluronic acid, and polyethylene glycol using fluorescence correlation spectroscopy and thioflavin T assays. Our results reveal that all hydrogels tested were associated with enhanced Aβ aggregation and Aβ cytotoxicity attenuation. We suggest that confinement itself imparts a profound effect, possibly by stabilizing Aβ structures and shifting the aggregate equilibrium toward larger species. If this phenomenon of altered protein aggregation in 3D hydrogels can be generalized to other contexts including the in vivo environment, it may be necessary to reevaluate aspects of protein aggregation disease models used for drug discovery.
Collapse
Affiliation(s)
- Laura
W. Simpson
- Department
of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Eng 314, Baltimore, Maryland 21250, United States
| | - Gregory L. Szeto
- Department
of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Eng 314, Baltimore, Maryland 21250, United States
- Marlene
and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, 22 S Greene Street, Baltimore, Maryland 21201, United
States
| | - Hacene Boukari
- Division
of Physical and Computational Sciences, Delaware State University, 1200 N. Dupont Highway, Dover, Delaware 19901, United States
| | - Theresa A. Good
- Division
of Molecular and Cellular Biosciences, National
Science Foundation, 2415 Eisenhower Avenue, E 12485, Alexandria, Virginia 22314, United States
| | - Jennie B. Leach
- Department
of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Eng 314, Baltimore, Maryland 21250, United States
| |
Collapse
|
20
|
Collagen hydrogel confinement of Amyloid-β (Aβ) accelerates aggregation and reduces cytotoxic effects. Acta Biomater 2020; 112:164-173. [PMID: 32464268 DOI: 10.1016/j.actbio.2020.05.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is associated with the accumulation of amyloid-β (Aβ), a peptide whose aggregation has been associated with neurotoxicity. Drugs targeting Aβ have shown great promise in 2D in vitro models and mouse models, yet preclinical and clinical trials for AD have been highly disappointing. We propose that current in vitro culture systems for discovering and developing AD drugs have significant limitations; specifically, that Aβ aggregation is vastly different in these 2D cultures carried out on flat plastic or glass substrates vs. in a 3D environment, such as brain tissue, where Aβ confinement alters aggregation kinetics and thermodynamics. In this work, we identified attenuation of Aβ cytotoxicity in 3D hydrogel culture compared to 2D cell culture. We investigated Aβ structure and aggregation in solution vs. hydrogel using Transmission Electron Microscopy (TEM), Fluorescence Correlation Spectroscopy (FCS), and Thioflavin T (ThT) assays. Our results reveal that the equilibrium is shifted to stable extended β-sheet (ThT positive) aggregates in hydrogels and away from the relatively unstable/unstructured presumed toxic oligomeric Aβ species in solution. Volume exclusion imparted by hydrogel confinement stabilizes unfolded, presumably toxic species, promoting stable extended β-sheet fibrils. STATEMENT OF SIGNIFICANCE: Alzheimer's disease (AD) is a devastating disease and has been studied for over 100 years. Yet, no cure exists and only 5 prescription drugs are FDA-approved to temporarily treat the AD symptoms of declining brain functions related to thinking and memory. Why don't we have more effective treatments to cure AD or relieve AD symptoms? We propose that current culture methods based upon cells cultured on flat, stiff substrates have significant limitations for discovering and developing AD drugs. This study provides strong evidence that AD drugs should be tested in 3D culture systems as a step along the development pathway towards new, more effective drugs to treat AD.
Collapse
|
21
|
Ovsepian SV, Horacek J, O'Leary VB, Hoschl C. The Ups and Downs of BACE1: Walking a Fine Line between Neurocognitive and Other Psychiatric Symptoms of Alzheimer's Disease. Neuroscientist 2020; 27:222-234. [PMID: 32713260 DOI: 10.1177/1073858420940943] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although neurocognitive deficit is the best-recognized indicator of Alzheimer's disease (AD), psychotic and other noncognitive symptoms are the prime cause of institutionalization. BACE1 is the rate-limiting enzyme in the production of Aβ of AD, and one of the promising therapeutic targets in countering cognitive decline and amyloid pathology. Changes in BACE1 activity have also emerged to cause significant noncognitive neuropsychiatric symptoms and impairments of circadian rhythms, as evident from clinical trials and reports in transgenic models. In this study, we consider key characteristics of BACE1 with its contribution to neurocognitive deficit and other psychiatric symptoms of AD. We argue that a growing list of noncognitive mental impairments related to pharmacological modulation of BACE1 might present a major obstacle in clinical translation of emerging therapeutic leads targeting this protease. The adverse effects of BACE1 inhibition on mental health call for a revision of treatment strategies that assume indiscriminate inhibition of this key protease, and stress the need for further mechanistic and translational studies.
Collapse
Affiliation(s)
- Saak V Ovsepian
- National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.,International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland
| | - Jiri Horacek
- National Institute of Mental Health, Klecany, Czech Republic
| | - Valerie B O'Leary
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Cyril Hoschl
- National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
22
|
Simpson LW, Good TA, Leach JB. Protein folding and assembly in confined environments: Implications for protein aggregation in hydrogels and tissues. Biotechnol Adv 2020; 42:107573. [PMID: 32512220 DOI: 10.1016/j.biotechadv.2020.107573] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 05/03/2020] [Accepted: 05/30/2020] [Indexed: 12/20/2022]
Abstract
In the biological milieu of a cell, soluble crowding molecules and rigid confined environments strongly influence whether the protein is properly folded, intrinsically disordered proteins assemble into distinct phases, or a denatured or aggregated protein species is favored. Such crowding and confinement factors act to exclude solvent volume from the protein molecules, resulting in an increased local protein concentration and decreased protein entropy. A protein's structure is inherently tied to its function. Examples of processes where crowding and confinement may strongly influence protein function include transmembrane protein dimerization, enzymatic activity, assembly of supramolecular structures (e.g., microtubules), nuclear condensates containing transcriptional machinery, protein aggregation in the contexts of disease and protein therapeutics. Historically, most protein structures have been determined from pure, dilute protein solutions or pure crystals. However, these are not the environments in which these proteins function. Thus, there has been an increased emphasis on analyzing protein structure and dynamics in more "in vivo-like" environments. Complex in vitro models using hydrogel scaffolds to study proteins may better mimic features of the in vivo environment. Therefore, analytical techniques need to be optimized for real-time analysis of proteins within hydrogel scaffolds.
Collapse
Affiliation(s)
- Laura W Simpson
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Theresa A Good
- Division of Molecular and Cellular Biosciences, National Science Foundation, 2415 Eisenhower Ave, Alexandria, VA 22314, USA
| | - Jennie B Leach
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
23
|
The molecular lifecycle of amyloid – Mechanism of assembly, mesoscopic organisation, polymorphism, suprastructures, and biological consequences. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1867:140257. [DOI: 10.1016/j.bbapap.2019.07.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/12/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022]
|
24
|
Shin Y, Choi SH, Kim E, Bylykbashi E, Kim JA, Chung S, Kim DY, Kamm RD, Tanzi RE. Blood-Brain Barrier Dysfunction in a 3D In Vitro Model of Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900962. [PMID: 31637161 PMCID: PMC6794630 DOI: 10.1002/advs.201900962] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/20/2019] [Indexed: 05/21/2023]
Abstract
Harmful materials in the blood are prevented from entering the healthy brain by a highly selective blood-brain barrier (BBB), and impairment of barrier function has been associated with a variety of neurological diseases. In Alzheimer's disease (AD), BBB breakdown has been shown to occur even before cognitive decline and brain pathology. To investigate the role of the cerebral vasculature in AD, a physiologically relevant 3D human neural cell culture microfluidic model is developed having a brain endothelial cell monolayer with a BBB-like phenotype. This model is shown to recapitulate several key aspects of BBB dysfunction observed in AD patients: increased BBB permeability, decreased expression of claudin-1, claudin-5, and VE-cadherin, increased expression of matrix-metalloproteinase-2 and reactive oxygen species, and deposition of β-amyloid (Aβ) peptides at the vascular endothelium. Thus, it provides a well-controlled platform for investigating BBB function as well as for screening of new drugs that need to pass the BBB to gain access to neural tissues.
Collapse
Affiliation(s)
- Yoojin Shin
- Department of Mechanical EngineeringMassachusetts Institute of Technology500 Technology Square, MIT Building, Room NE47‐321CambridgeMA02139USA
| | - Se Hoon Choi
- Genetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseDepartment of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Eunhee Kim
- Genetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseDepartment of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Enjana Bylykbashi
- Genetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseDepartment of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Jeong Ah Kim
- Biomedical Omics GroupKorea Basic Science InstituteCheongju28119Republic of Korea
- Department of Bio‐Analytical ScienceUniversity of Science and TechnologyDaejeon34113Republic of Korea
| | - Seok Chung
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- School of Mechanical EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Doo Yeon Kim
- Genetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseDepartment of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Roger D. Kamm
- Department of Mechanical EngineeringMassachusetts Institute of Technology500 Technology Square, MIT Building, Room NE47‐321CambridgeMA02139USA
- Department of Biological EngineeringMassachusetts Institute of Technology500 Technology Square, MIT Building, Room NE47‐321CambridgeMA02139USA
- Singapore‐MIT Alliance for Research & Technology (SMART)BioSystems and Micromechanics (BioSyM)Singapore138602Singapore
| | - Rudolph E. Tanzi
- Genetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseDepartment of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| |
Collapse
|
25
|
Abstract
The brain is the most complex organ of the body, and many pathological processes underlying various brain disorders are poorly understood. Limited accessibility hinders observation of such processes in the in vivo brain, and experimental freedom is often insufficient to enable informative manipulations. In vitro preparations (brain slices or cultures of dissociated neurons) offer much better accessibility and reduced complexity and have yielded valuable new insights into various brain disorders. Both types of preparations have their advantages and limitations with regard to lifespan, preservation of in vivo brain structure, composition of cell types, and the link to behavioral outcome is often unclear in in vitro models. While these limitations hamper general usage of in vitro preparations to study, e.g., brain development, in vitro preparations are very useful to study neuronal and synaptic functioning under pathologic conditions. This chapter addresses several brain disorders, focusing on neuronal and synaptic functioning, as well as network aspects. Recent progress in the fields of brain circulation disorders, excitability disorders, and memory disorders will be discussed, as well as limitations of current in vitro models.
Collapse
|
26
|
Kuznetsov IA, Kuznetsov AV. Simulating the effect of formation of amyloid plaques on aggregation of tau protein. Proc Math Phys Eng Sci 2018; 474:20180511. [PMID: 30602936 PMCID: PMC6304026 DOI: 10.1098/rspa.2018.0511] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022] Open
Abstract
In this paper, we develop a mathematical model that enables the investigation of the production and intracellular transport of amyloid precursor protein (APP) and tau protein in a neuron. We also investigate the aggregation of APP fragments into amyloid-β (Aβ) as well as tau aggregation into tau oligomers and neurofibrillary tangles. Using the developed model, we investigate how Aβ aggregation can influence tau transport and aggregation in both the soma and the axon. We couple the Aβ and tau agglomeration processes by assuming that the value of the kinetic constant that describes the autocatalytic growth (self-replication) reaction step of tau aggregation is proportional to the Aβ concentration. The model predicts that APP and tau are distributed differently in the axon. While APP has a uniform distribution along the axon, tau's concentration first decreases and then increases towards the synapse. Aβ is uniformly produced along the axon while misfolded tau protein is mostly produced in the proximal axon. The number of Aβ and tau polymers originating from the axon is much smaller than the number of Aβ and tau polymers originating from the soma. The rate of production of misfolded tau polymers depends on how strongly their production is facilitated by Aβ.
Collapse
Affiliation(s)
- I. A. Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - A. V. Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA
| |
Collapse
|
27
|
Toglia P, Demuro A, Mak DOD, Ullah G. Data-driven modeling of mitochondrial dysfunction in Alzheimer's disease. Cell Calcium 2018; 76:23-35. [PMID: 30248575 DOI: 10.1016/j.ceca.2018.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/02/2018] [Accepted: 09/03/2018] [Indexed: 02/06/2023]
Abstract
Intracellular accumulation of oligomeric forms of β amyloid (Aβ) are now believed to play a key role in the earliest phase of Alzheimer's disease (AD) as their rise correlates well with the early symptoms of the disease. Extensive evidence points to impaired neuronal Ca2+ homeostasis as a direct consequence of the intracellular Aβ oligomers. However, little is known about the downstream effects of the resulting Ca2+ rise on the many intracellular Ca2+-dependent pathways. Here we use multiscale modeling in conjunction with patch-clamp electrophysiology of single inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) and fluorescence imaging of whole-cell Ca2+ response, induced by exogenously applied intracellular Aβ42 oligomers to show that Aβ42 inflicts cytotoxicity by impairing mitochondrial function. Driven by patch-clamp experiments, we first model the kinetics of IP3R, which is then extended to build a model for the whole-cell Ca2+ signals. The whole-cell model is then fitted to fluorescence signals to quantify the overall Ca2+ release from the endoplasmic reticulum by intracellular Aβ42 oligomers through G-protein-mediated stimulation of IP3 production. The estimated IP3 concentration as a function of intracellular Aβ42 content together with the whole-cell model allows us to show that Aβ42 oligomers impair mitochondrial function through pathological Ca2+ uptake and the resulting reduced mitochondrial inner membrane potential, leading to an overall lower ATP and increased production of reactive oxygen species and H2O2. We further show that mitochondrial function can be restored by the addition of Ca2+ buffer EGTA, in accordance with the observed abrogation of Aβ42 cytotoxicity by EGTA in our live cells experiments.
Collapse
Affiliation(s)
- Patrick Toglia
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
| | - Angelo Demuro
- Department of Neurobiology and Behavior and Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Don-On Daniel Mak
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
28
|
Newton AJH, McDougal RA, Hines ML, Lytton WW. Using NEURON for Reaction-Diffusion Modeling of Extracellular Dynamics. Front Neuroinform 2018; 12:41. [PMID: 30042670 PMCID: PMC6049079 DOI: 10.3389/fninf.2018.00041] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 06/12/2018] [Indexed: 11/13/2022] Open
Abstract
Development of credible clinically-relevant brain simulations has been slowed due to a focus on electrophysiology in computational neuroscience, neglecting the multiscale whole-tissue modeling approach used for simulation in most other organ systems. We have now begun to extend the NEURON simulation platform in this direction by adding extracellular modeling. The extracellular medium of neural tissue is an active medium of neuromodulators, ions, inflammatory cells, oxygen, NO and other gases, with additional physiological, pharmacological and pathological agents. These extracellular agents influence, and are influenced by, cellular electrophysiology, and cellular chemophysiology-the complex internal cellular milieu of second-messenger signaling and cascades. NEURON's extracellular reaction-diffusion is supported by an intuitive Python-based where/who/what command sequence, derived from that used for intracellular reaction diffusion, to support coarse-grained macroscopic extracellular models. This simulation specification separates the expression of the conceptual model and parameters from the underlying numerical methods. In the volume-averaging approach used, the macroscopic model of tissue is characterized by free volume fraction-the proportion of space in which species are able to diffuse, and tortuosity-the average increase in path length due to obstacles. These tissue characteristics can be defined within particular spatial regions, enabling the modeler to account for regional differences, due either to intrinsic organization, particularly gray vs. white matter, or to pathology such as edema. We illustrate simulation development using spreading depression, a pathological phenomenon thought to play roles in migraine, epilepsy and stroke. Simulation results were verified against analytic results and against the extracellular portion of the simulation run under FiPy. The creation of this NEURON interface provides a pathway for interoperability that can be used to automatically export this class of models into complex intracellular/extracellular simulations and future cross-simulator standardization.
Collapse
Affiliation(s)
- Adam J. H. Newton
- Department of Neuroscience, Yale University, New Haven, CT, United States
- SUNY Downstate Medical Center, The State University of New York, New York, NY, United States
| | - Robert A. McDougal
- Department of Neuroscience, Yale University, New Haven, CT, United States
- Center for Medical Informatics, Yale University, New Haven, CT, United States
| | - Michael L. Hines
- Department of Neuroscience, Yale University, New Haven, CT, United States
| | - William W. Lytton
- SUNY Downstate Medical Center, The State University of New York, New York, NY, United States
- Neurology, Kings County Hospital Center, Brooklyn, NY, United States
| |
Collapse
|
29
|
Rammes G, Mattusch C, Wulff M, Seeser F, Kreuzer M, Zhu K, Deussing JM, Herms J, Parsons CG. Involvement of GluN2B subunit containing N-methyl-d-aspartate (NMDA) receptors in mediating the acute and chronic synaptotoxic effects of oligomeric amyloid-beta (Aβ) in murine models of Alzheimer's disease (AD). Neuropharmacology 2017; 123:100-115. [PMID: 28174113 DOI: 10.1016/j.neuropharm.2017.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/17/2017] [Accepted: 02/03/2017] [Indexed: 01/05/2023]
Abstract
To elucidate whether a permanent reduction of the GluN2B subunit affects the pathology of Alzheimer's disease (AD), we cross-bred mice heterozygous for GluN2B receptors in the forebrain (hetGluN2B) with a mouse model for AD carrying a mutated amyloid precursor protein with the Swedish and Arctic mutation (mAPP) resulting in a hetGluN2B/mAPP transgenic. By means of voltage-sensitive dye imaging (VSDI) in the di-synaptic hippocampal pathway and the recording of field excitatory postsynaptic potentials (fEPSPs), hippocampal slices of all genotypes (WT, hetGluN2B, mAPP and hetGluN2B/mAPP, age 9-18 months) were tested for spatiotemporal activity propagation and long-term potentiation (LTP) induction. CA1-LTP induced by high frequency stimulation (HFS; 100 Hz/1s) was not different in all genotypes. Aβ1-42 (50 nM)-application reduced potentiation of fEPSP in WT and hetGluN2B/mAPP mice, LTP in mAPP and hetGluN2B mice was not affected. For VSDI a fast depolarization signal was evoked in the granule cell layer and propagation was analysed in hippocampal CA3 and CA1 region before and after theta stimulation (100pulses/5 Hz). LTP was not significantly different between all genotypes. In mAPP mice θ-stim produced an epileptiform activity reflected in a pronounced prolongation of the FDS compared to the other genotypes. In slices of hetGluN2B/mAPP and GluN2B mice, however, these parameters were similar to WT mice indicating a reversal effect of the attenuated GluN2B expression. The induction of a hetGluN2B mutation in the mAPP reversed some pathophysiological changes on hippocampal LTP and provide further evidence for the involvement of the glutamatergic system in AD and emphasize the GluN2B subunit as a potential target for AD treatment.
Collapse
Affiliation(s)
- Gerhard Rammes
- Department of Anaesthesiology, Technische Universität München, Munich, Germany; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.
| | - Corinna Mattusch
- Department of Anaesthesiology, Technische Universität München, Munich, Germany
| | - Matthias Wulff
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Franziska Seeser
- Department of Anaesthesiology, Technische Universität München, Munich, Germany
| | - Matthias Kreuzer
- Department of Anaesthesiology, Technische Universität München, Munich, Germany
| | - Kaichuan Zhu
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Chris G Parsons
- Non-Clinical Science, Merz Pharmaceuticals GmbH, Frankfurt am Main, Germany
| |
Collapse
|
30
|
Hao W, Friedman A. Mathematical model on Alzheimer's disease. BMC SYSTEMS BIOLOGY 2016; 10:108. [PMID: 27863488 PMCID: PMC5116206 DOI: 10.1186/s12918-016-0348-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 10/25/2016] [Indexed: 12/21/2022]
Abstract
Background Alzheimer disease (AD) is a progressive neurodegenerative disease that destroys memory and cognitive skills. AD is characterized by the presence of two types of neuropathological hallmarks: extracellular plaques consisting of amyloid β-peptides and intracellular neurofibrillary tangles of hyperphosphorylated tau proteins. The disease affects 5 million people in the United States and 44 million world-wide. Currently there is no drug that can cure, stop or even slow the progression of the disease. If no cure is found, by 2050 the number of alzheimer’s patients in the U.S. will reach 15 million and the cost of caring for them will exceed $ 1 trillion annually. Results The present paper develops a mathematical model of AD that includes neurons, astrocytes, microglias and peripheral macrophages, as well as amyloid β aggregation and hyperphosphorylated tau proteins. The model is represented by a system of partial differential equations. The model is used to simulate the effect of drugs that either failed in clinical trials, or are currently in clinical trials. Conclusions Based on these simulations it is suggested that combined therapy with TNF- α inhibitor and anti amyloid β could yield significant efficacy in slowing the progression of AD.
Collapse
Affiliation(s)
- Wenrui Hao
- Department of Mathematics, The Penn State University, University Park, 16802, PA, USA.
| | - Avner Friedman
- Mathematical Biosciences Institute & Department of Mathematics, The Ohio State University, Columbus, 43210, OH, USA
| |
Collapse
|
31
|
Hascup KN, Hascup ER. Soluble Amyloid-β42 Stimulates Glutamate Release through Activation of the α7 Nicotinic Acetylcholine Receptor. J Alzheimers Dis 2016; 53:337-47. [DOI: 10.3233/jad-160041] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Kevin N. Hascup
- Department of Neurology, Center for Alzheimer’s Disease and Related Disorders, Neurosciences Institute, Center for Integrated Research in Cognitive & Neural Sciences, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R. Hascup
- Department of Neurology, Center for Alzheimer’s Disease and Related Disorders, Neurosciences Institute, Center for Integrated Research in Cognitive & Neural Sciences, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
32
|
Novotny R, Langer F, Mahler J, Skodras A, Vlachos A, Wegenast-Braun BM, Kaeser SA, Neher JJ, Eisele YS, Pietrowski MJ, Nilsson KPR, Deller T, Staufenbiel M, Heimrich B, Jucker M. Conversion of Synthetic Aβ to In Vivo Active Seeds and Amyloid Plaque Formation in a Hippocampal Slice Culture Model. J Neurosci 2016; 36:5084-93. [PMID: 27147660 PMCID: PMC6601857 DOI: 10.1523/jneurosci.0258-16.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/21/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED The aggregation of amyloid-β peptide (Aβ) in brain is an early event and hallmark of Alzheimer's disease (AD). We combined the advantages of in vitro and in vivo approaches to study cerebral β-amyloidosis by establishing a long-term hippocampal slice culture (HSC) model. While no Aβ deposition was noted in untreated HSCs of postnatal Aβ precursor protein transgenic (APP tg) mice, Aβ deposition emerged in HSCs when cultures were treated once with brain extract from aged APP tg mice and the culture medium was continuously supplemented with synthetic Aβ. Seeded Aβ deposition was also observed under the same conditions in HSCs derived from wild-type or App-null mice but in no comparable way when HSCs were fixed before cultivation. Both the nature of the brain extract and the synthetic Aβ species determined the conformational characteristics of HSC Aβ deposition. HSC Aβ deposits induced a microglia response, spine loss, and neuritic dystrophy but no obvious neuron loss. Remarkably, in contrast to in vitro aggregated synthetic Aβ, homogenates of Aβ deposits containing HSCs induced cerebral β-amyloidosis upon intracerebral inoculation into young APP tg mice. Our results demonstrate that a living cellular environment promotes the seeded conversion of synthetic Aβ into a potent in vivo seeding-active form. SIGNIFICANCE STATEMENT In this study, we report the seeded induction of Aβ aggregation and deposition in long-term hippocampal slice cultures. Remarkably, we find that the biological activities of the largely synthetic Aβ aggregates in the culture are very similar to those observed in vivo This observation is the first to show that potent in vivo seeding-active Aβ aggregates can be obtained by seeded conversion of synthetic Aβ in a living (wild-type) cellular environment.
Collapse
Affiliation(s)
- Renata Novotny
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen D-72076, Germany, DZNE, German Center for Neurodegenerative Diseases, Tübingen D-72076, Germany, Graduate School for Cellular and Molecular Neuroscience, University of Tübingen, Tübingen D-72076, Germany
| | - Franziska Langer
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen D-72076, Germany, DZNE, German Center for Neurodegenerative Diseases, Tübingen D-72076, Germany
| | - Jasmin Mahler
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen D-72076, Germany, DZNE, German Center for Neurodegenerative Diseases, Tübingen D-72076, Germany, Graduate School for Cellular and Molecular Neuroscience, University of Tübingen, Tübingen D-72076, Germany
| | - Angelos Skodras
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen D-72076, Germany, DZNE, German Center for Neurodegenerative Diseases, Tübingen D-72076, Germany
| | - Andreas Vlachos
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt/Main D-60590, Germany
| | - Bettina M Wegenast-Braun
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen D-72076, Germany, DZNE, German Center for Neurodegenerative Diseases, Tübingen D-72076, Germany
| | - Stephan A Kaeser
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen D-72076, Germany, DZNE, German Center for Neurodegenerative Diseases, Tübingen D-72076, Germany
| | - Jonas J Neher
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen D-72076, Germany, DZNE, German Center for Neurodegenerative Diseases, Tübingen D-72076, Germany
| | - Yvonne S Eisele
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen D-72076, Germany, DZNE, German Center for Neurodegenerative Diseases, Tübingen D-72076, Germany
| | - Marie J Pietrowski
- Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg D-79104, Germany, and
| | - K Peter R Nilsson
- Department of Chemistry, IFM, Linköping University, Linköping SE-581 83, Sweden
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt/Main D-60590, Germany
| | - Matthias Staufenbiel
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen D-72076, Germany, DZNE, German Center for Neurodegenerative Diseases, Tübingen D-72076, Germany
| | - Bernd Heimrich
- Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg D-79104, Germany, and
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen D-72076, Germany, DZNE, German Center for Neurodegenerative Diseases, Tübingen D-72076, Germany,
| |
Collapse
|
33
|
Abstract
Amyloid β (Aβ) is thought to play an important role in the pathogenesis of Alzheimer's disease. Aβ may exert its neurotoxic effects via multiple mechanisms and in particular through degradation of excitatory synaptic transmission associated with impaired synaptic plasticity. In contrast, much less is known about Aβ effects at inhibitory synapses. This study investigates the impact of acute Aβ1-42 application on GABAergic synaptic transmission in rat somatosensory cortex in vitro. Whole-cell voltage-clamp recordings were obtained from layer V pyramidal cells, and monosynaptic GABA(A) receptor-mediated IPSCs were elicited. Bath-applied Aβ (1 μm) depressed the IPSCs on average to 60% of control, whereas a reversed sequence control peptide was ineffective. Paired-pulse stimuli indicated a postsynaptic site of action. This was further corroborated by a decreased postsynaptic responsiveness to local puffs of the GABAA receptor agonist isoguvacine. The Aβ-induced IPSC decline could be prevented with intracellular applications of p4, a blocker of GABA(A) receptor internalization. It is concluded that Aβ weakens synaptic inhibition via downregulation of GABA(A) receptors.
Collapse
|
34
|
Iturria-Medina Y, Evans AC. On the central role of brain connectivity in neurodegenerative disease progression. Front Aging Neurosci 2015; 7:90. [PMID: 26052284 PMCID: PMC4439541 DOI: 10.3389/fnagi.2015.00090] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 05/01/2015] [Indexed: 12/12/2022] Open
Abstract
Increased brain connectivity, in all its variants, is often considered an evolutionary advantage by mediating complex sensorimotor function and higher cognitive faculties. Interaction among components at all spatial scales, including genes, proteins, neurons, local neuronal circuits and macroscopic brain regions, are indispensable for such vital functions. However, a growing body of evidence suggests that, from the microscopic to the macroscopic levels, such connections might also be a conduit for in intra-brain disease spreading. For instance, cell-to-cell misfolded proteins (MP) transmission and neuronal toxicity are prominent connectivity-mediated factors in aging and neurodegeneration. This article offers an overview of connectivity dysfunctions associated with neurodegeneration, with a specific focus on how these may be central to both normal aging and the neuropathologic degenerative progression.
Collapse
Affiliation(s)
- Yasser Iturria-Medina
- Montreal Neurological Institute Montreal, QC, Canada ; Ludmer Center for NeuroInformatics and Mental Health Montreal, QC, Canada
| | - Alan C Evans
- Montreal Neurological Institute Montreal, QC, Canada ; Ludmer Center for NeuroInformatics and Mental Health Montreal, QC, Canada
| |
Collapse
|
35
|
Tamagnini F, Scullion S, Brown JT, Randall AD. Intrinsic excitability changes induced by acute treatment of hippocampal CA1 pyramidal neurons with exogenous amyloid β peptide. Hippocampus 2015; 25:786-97. [PMID: 25515596 PMCID: PMC4791149 DOI: 10.1002/hipo.22403] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2014] [Indexed: 12/17/2022]
Abstract
Accumulation of beta‐amyloid (Aβ) peptides in the human brain is a canonical pathological hallmark of Alzheimer's disease (AD). Recent work in Aβ‐overexpressing transgenic mice indicates that increased brain Aβ levels can be associated with aberrant epileptiform activity. In line with this, such mice can also exhibit altered intrinsic excitability (IE) of cortical and hippocampal neurons: these observations may relate to the increased prevalence of seizures in AD patients. In this study, we examined what changes in IE are produced in hippocampal CA1 pyramidal cells after 2–5 h treatment with an oligomeric preparation of synthetic human Aβ 1–42 peptide. Whole cell current clamp recordings were compared between Aβ‐(500 nM) and vehicle‐(DMSO 0.05%) treated hippocampal slices obtained from mice. The soluble Aβ treatment did not produce alterations in sub‐threshold intrinsic properties, including membrane potential, input resistance, and hyperpolarization activated “sag”. Similarly, no changes were noted in the firing profile evoked by 500 ms square current supra‐threshold stimuli. However, Aβ 500 nM treatment resulted in the hyperpolarization of the action potential (AP) threshold. In addition, treatment with Aβ at 500 nM depressed the after‐hyperpolarization that followed both a single AP or 50 Hz trains of a number of APs between 5 and 25. These data suggest that acute exposure to soluble Aβ oligomers affects IE properties of CA1 pyramidal neurons differently from outcomes seen in transgenic models of amyloidopathy. However, in both chronic and acute models, the IE changes are toward hyperexcitability, reinforcing the idea that amyloidopathy and increased incidence in seizures might be causally related in AD patients. © 2014 The Authors Hippocampus Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Francesco Tamagnini
- Medical School, University of Exeter, Hatherly Building, Streatham Campus, Exeter, EX4 4PS, United Kingdom.,School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, BS8 1TD, United Kingdom
| | - Sarah Scullion
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, BS8 1TD, United Kingdom
| | - Jon T Brown
- Medical School, University of Exeter, Hatherly Building, Streatham Campus, Exeter, EX4 4PS, United Kingdom.,School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, BS8 1TD, United Kingdom
| | - Andrew D Randall
- Medical School, University of Exeter, Hatherly Building, Streatham Campus, Exeter, EX4 4PS, United Kingdom.,School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, BS8 1TD, United Kingdom
| |
Collapse
|
36
|
Aβ42 oligomers selectively disrupt neuronal calcium release. Neurobiol Aging 2015; 36:877-85. [PMID: 25453559 DOI: 10.1016/j.neurobiolaging.2014.10.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 10/09/2014] [Accepted: 10/14/2014] [Indexed: 12/30/2022]
|
37
|
Iturria-Medina Y, Sotero RC, Toussaint PJ, Evans AC, and the Alzheimer's Disease Neuroimaging Initiative. Epidemic spreading model to characterize misfolded proteins propagation in aging and associated neurodegenerative disorders. PLoS Comput Biol 2014; 10:e1003956. [PMID: 25412207 PMCID: PMC4238950 DOI: 10.1371/journal.pcbi.1003956] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 10/01/2014] [Indexed: 12/20/2022] Open
Abstract
Misfolded proteins (MP) are a key component in aging and associated neurodegenerative disorders. For example, misfolded Amyloid-ß (Aß) and tau proteins are two neuropathogenic hallmarks of Alzheimer's disease. Mechanisms underlying intra-brain MP propagation/deposition remain essentially uncharacterized. Here, is introduced an epidemic spreading model (ESM) for MP dynamics that considers propagation-like interactions between MP agents and the brain's clearance response across the structural connectome. The ESM reproduces advanced Aß deposition patterns in the human brain (explaining 46∼56% of the variance in regional Aß loads, in 733 subjects from the ADNI database). Furthermore, this model strongly supports a) the leading role of Aß clearance deficiency and early Aß onset age during Alzheimer's disease progression, b) that effective anatomical distance from Aß outbreak region explains regional Aß arrival time and Aß deposition likelihood, c) the multi-factorial impact of APOE e4 genotype, gender and educational level on lifetime intra-brain Aß propagation, and d) the modulatory impact of Aß propagation history on tau proteins concentrations, supporting the hypothesis of an interrelated pathway between Aß pathophysiology and tauopathy. To our knowledge, the ESM is the first computational model highlighting the direct link between structural brain networks, production/clearance of pathogenic proteins and associated intercellular transfer mechanisms, individual genetic/demographic properties and clinical states in health and disease. In sum, the proposed ESM constitutes a promising framework to clarify intra-brain region to region transference mechanisms associated with aging and neurodegenerative disorders. Misfolded proteins (MP) mechanisms are a characteristic pathogenic feature of most prevalent human neurodegenerative diseases, such as Alzheimer's disease (AD). Characterizing the mechanisms underlying intra-brain MP propagation and deposition still constitutes a major challenge. Here, we hypothesize that these complex mechanisms can be accurately described by epidemic spreading-like interactions between infectious-like agents (MP) and the brain's MP clearance response, which are constrained by the brain's connectional architecture. Consequently, we have developed a stochastic epidemic spreading model (ESM) of MP propagation/deposition that allows for reconstructing individual lifetime histories of intra-brain MP propagation, and the subsequent analysis of factors that promote propagation/deposition (e.g., MP production and clearance). Using 733 individual PET Amyloid-ß (Aß) datasets, we show that ESM explains advanced Aß deposition patterns in healthy and diseased (AD) brains. More importantly, it offers new avenues for our understanding of the mechanisms underlying MP mediated disorders. For instance, the results strongly support the growing body of evidence suggesting the leading role of a reduced Aβ clearance on AD progression and the modulatory impact of Aß mechanisms on tau proteins concentrations, which could imply a turning point for associated therapeutic mitigation strategies.
Collapse
Affiliation(s)
| | | | | | - Alan C. Evans
- Montreal Neurological Institute, Montreal, Quebec, Canada
- * E-mail: (YIM); (ACE)
| | | |
Collapse
|
38
|
McCarthy RC, Park YH, Kosman DJ. sAPP modulates iron efflux from brain microvascular endothelial cells by stabilizing the ferrous iron exporter ferroportin. EMBO Rep 2014; 15:809-15. [PMID: 24867889 DOI: 10.15252/embr.201338064] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A sequence within the E2 domain of soluble amyloid precursor protein (sAPP) stimulates iron efflux. This activity has been attributed to a ferroxidase activity suggested for this motif. We demonstrate that the stimulation of efflux supported by this peptide and by sAPPα is due to their stabilization of the ferrous iron exporter, ferroportin (Fpn), in the plasma membrane of human brain microvascular endothelial cells (hBMVEC). The peptide does not bind ferric iron explaining why it does not and thermodynamically cannot promote ferrous iron autoxidation. This peptide specifically pulls Fpn down from the plasma membrane of hBMVEC; based on these results, FTP, for ferroportin-targeting peptide, correctly identifies the function of this peptide. The data suggest that in stabilizing Fpn via the targeting due to the FTP sequence, sAPP will increase the flux of iron into the cerebral interstitium. This inference correlates with the observation of significant iron deposition in the amyloid plaques characteristic of Alzheimer's disease.
Collapse
Affiliation(s)
- Ryan C McCarthy
- Department of Biochemistry, School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| | - Yun-Hee Park
- Department of Biochemistry, School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| | - Daniel J Kosman
- Department of Biochemistry, School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
39
|
Lee HH, Choi TS, Lee SJC, Lee JW, Park J, Ko YH, Kim WJ, Kim K, Kim HI. Supramolecular Inhibition of Amyloid Fibrillation by Cucurbit[7]uril. Angew Chem Int Ed Engl 2014; 53:7461-5. [DOI: 10.1002/anie.201402496] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 03/26/2014] [Indexed: 11/11/2022]
|
40
|
Lee HH, Choi TS, Lee SJC, Lee JW, Park J, Ko YH, Kim WJ, Kim K, Kim HI. Supramolecular Inhibition of Amyloid Fibrillation by Cucurbit[7]uril. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201402496] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
41
|
Briggs CA, Schneider C, Richardson JC, Stutzmann GE. β amyloid peptide plaques fail to alter evoked neuronal calcium signals in APP/PS1 Alzheimer's disease mice. Neurobiol Aging 2013; 34:1632-43. [PMID: 23337342 DOI: 10.1016/j.neurobiolaging.2012.12.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 12/04/2012] [Accepted: 12/18/2012] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial disorder of unknown etiology. Mechanistically, beta amyloid peptides (Aβ) and elevated Ca(2+) have been implicated as proximal and likely interactive features of the disease process. We tested the hypothesis that proximity to Aβ plaque might exacerbate activity-dependent neuronal Ca(2+) signaling in hippocampal pyramidal neurons from APPSWE/PS1M146V mice. Using combined approaches of whole cell patch clamp recording and 2-photon imaging of neuronal Ca(2+) signals with thioflavin-S plaque labeling in hippocampal slices, we found no correlation between thioflavin-S labeled Aβ plaque proximity and Ca(2+) responses triggered by ryanodine receptor (RyR) activation or action potentials in either dendrites or somata of AD mice, regardless of age. Baseline and RyR-stimulated spontaneous excitatory postsynaptic potentials also showed little difference in relation to Aβ plaque proximity. Consistent with previous studies, RyR-evoked Ca(2+) release in APPSWE/PS1M146V mice was greater than in nontransgenic controls. Within the soma, RyR-evoked Ca(2+) release was elevated in older APPSWE/PS1M146V mice compared with younger APPSWE/PS1M146V mice, but was still independent of plaque proximity. The results indicate that early Ca(2+) signaling disruptions can become yet more severe with age through mechanisms independent of Aβ plaques, suggesting that alternative pathogenic mechanisms might contribute to AD-associated dysfunction.
Collapse
Affiliation(s)
- Clark A Briggs
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | | | | | | |
Collapse
|
42
|
Tang Y, Scott DA, Das U, Edland SD, Radomski K, Koo EH, Roy S. Early and selective impairments in axonal transport kinetics of synaptic cargoes induced by soluble amyloid β-protein oligomers. Traffic 2012; 13:681-93. [PMID: 22309053 DOI: 10.1111/j.1600-0854.2012.01340.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Revised: 02/02/2012] [Accepted: 02/06/2012] [Indexed: 11/28/2022]
Abstract
The downstream targets of amyloid β (Aβ)-oligomers remain elusive. One hypothesis is that Aβ-oligomers interrupt axonal transport. Although previous studies have demonstrated Aβ-induced transport blockade, early effects of low-n soluble Aβ-oligomers on axonal transport remain unclear. Furthermore, the cargo selectivity for such deficits (if any) or the specific effects of Aβ on the motility kinetics of transported cargoes are also unknown. Toward this, we visualized axonal transport of vesicles in cultured hippocampal neurons treated with picomolar (pm) levels of cell-derived soluble Aβ-oligomers. We examined select cargoes thought to move as distinct organelles and established imaging parameters that allow organelle tracking with consistency and high fidelity - analyzing all data in a blinded fashion. Aβ-oligomers induced early and selective diminutions in velocities of synaptic cargoes but had no effect on mitochondrial motility, contrary to previous reports. These changes were N-methyl D-aspartate receptor/glycogen synthase kinase-3β dependent and reversible upon washout of the oligomers. Cluster-mode analyses reveal selective attenuations in faster-moving synaptic vesicles, suggesting possible decreases in cargo/motor associations, and biochemical experiments implicate tau phosphorylation in the process. Collectively, the data provide a biological basis for Aβ-induced axonal transport deficits.
Collapse
Affiliation(s)
- Yong Tang
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | | | | | |
Collapse
|