2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Faulty autolysosome acidification in Alzheimer’s disease mouse models induces autophagic build-up of Aβ in neurons, yielding senile plaques

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Autophagy is markedly impaired in Alzheimer’s disease (AD). Here we reveal unique autophagy dysregulation within neurons in five AD mouse models in vivo and identify its basis using a neuron-specific transgenic mRFP-eGFP-LC3 probe of autophagy and pH, multiplex confocal imaging and correlative light electron microscopy. Autolysosome acidification declines in neurons well before extracellular amyloid deposition, associated with markedly lowered vATPase activity and build-up of Aβ/APP-βCTF selectively within enlarged de-acidified autolysosomes. In more compromised yet still intact neurons, profuse Aβ-positive autophagic vacuoles (AVs) pack into large membrane blebs forming flower-like perikaryal rosettes. This unique pattern, termed PANTHOS (poisonous anthos (flower)), is also present in AD brains. Additional AVs coalesce into peri-nuclear networks of membrane tubules where fibrillar β-amyloid accumulates intraluminally. Lysosomal membrane permeabilization, cathepsin release and lysosomal cell death ensue, accompanied by microglial invasion. Quantitative analyses confirm that individual neurons exhibiting PANTHOS are the principal source of senile plaques in amyloid precursor protein AD models.

          Abstract

          Interrogation of neuronal autophagy in vivo in Alzheimerʼs disease mouse models identified deficient autolysosome acidification as the basis for extreme autophagic stress, yielding β-amyloid accumulation within intact neurons, which are the main source of senile plaques.

          Related collections

          Most cited references60

          • Record: found
          • Abstract: found
          • Article: not found

          Autophagy fights disease through cellular self-digestion.

          Autophagy, or cellular self-digestion, is a cellular pathway involved in protein and organelle degradation, with an astonishing number of connections to human disease and physiology. For example, autophagic dysfunction is associated with cancer, neurodegeneration, microbial infection and ageing. Paradoxically, although autophagy is primarily a protective process for the cell, it can also play a role in cell death. Understanding autophagy may ultimately allow scientists and clinicians to harness this process for the purpose of improving human health.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer's disease mutations: potential factors in amyloid plaque formation.

            Mutations in the genes for amyloid precursor protein (APP) and presenilins (PS1, PS2) increase production of beta-amyloid 42 (Abeta42) and cause familial Alzheimer's disease (FAD). Transgenic mice that express FAD mutant APP and PS1 overproduce Abeta42 and exhibit amyloid plaque pathology similar to that found in AD, but most transgenic models develop plaques slowly. To accelerate plaque development and investigate the effects of very high cerebral Abeta42 levels, we generated APP/PS1 double transgenic mice that coexpress five FAD mutations (5XFAD mice) and additively increase Abeta42 production. 5XFAD mice generate Abeta42 almost exclusively and rapidly accumulate massive cerebral Abeta42 levels. Amyloid deposition (and gliosis) begins at 2 months and reaches a very large burden, especially in subiculum and deep cortical layers. Intraneuronal Abeta42 accumulates in 5XFAD brain starting at 1.5 months of age (before plaques form), is aggregated (as determined by thioflavin S staining), and occurs within neuron soma and neurites. Some amyloid deposits originate within morphologically abnormal neuron soma that contain intraneuronal Abeta. Synaptic markers synaptophysin, syntaxin, and postsynaptic density-95 decrease with age in 5XFAD brain, and large pyramidal neurons in cortical layer 5 and subiculum are lost. In addition, levels of the activation subunit of cyclin-dependent kinase 5, p25, are elevated significantly at 9 months in 5XFAD brain, although an upward trend is observed by 3 months of age, before significant neurodegeneration or neuron loss. Finally, 5XFAD mice have impaired memory in the Y-maze. Thus, 5XFAD mice rapidly recapitulate major features of AD amyloid pathology and may be useful models of intraneuronal Abeta42-induced neurodegeneration and amyloid plaque formation.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Neuropathological alterations in Alzheimer disease.

              The neuropathological hallmarks of Alzheimer disease (AD) include "positive" lesions such as amyloid plaques and cerebral amyloid angiopathy, neurofibrillary tangles, and glial responses, and "negative" lesions such as neuronal and synaptic loss. Despite their inherently cross-sectional nature, postmortem studies have enabled the staging of the progression of both amyloid and tangle pathologies, and, consequently, the development of diagnostic criteria that are now used worldwide. In addition, clinicopathological correlation studies have been crucial to generate hypotheses about the pathophysiology of the disease, by establishing that there is a continuum between "normal" aging and AD dementia, and that the amyloid plaque build-up occurs primarily before the onset of cognitive deficits, while neurofibrillary tangles, neuron loss, and particularly synaptic loss, parallel the progression of cognitive decline. Importantly, these cross-sectional neuropathological data have been largely validated by longitudinal in vivo studies using modern imaging biomarkers such as amyloid PET and volumetric MRI.
                Bookmark

                Author and article information

                Contributors
                Ju-hyun.Lee@NKI.RFMH.ORG
                Ralph.Nixon@NKI.RFMH.ORG
                Journal
                Nat Neurosci
                Nat Neurosci
                Nature Neuroscience
                Nature Publishing Group US (New York )
                1097-6256
                1546-1726
                2 June 2022
                2 June 2022
                2022
                : 25
                : 6
                : 688-701
                Affiliations
                [1 ]GRID grid.250263.0, ISNI 0000 0001 2189 4777, Center for Dementia Research, , Nathan S. Kline Institute, ; Orangeburg, NY USA
                [2 ]GRID grid.137628.9, ISNI 0000 0004 1936 8753, Department of Psychiatry, , New York University Langone Health, ; New York, NY USA
                [3 ]GRID grid.418190.5, ISNI 0000 0001 2187 0556, Thermo Fisher Scientific, ; Hillsboro, OR USA
                [4 ]GRID grid.137628.9, ISNI 0000 0004 1936 8753, Departments of Biochemistry & Molecular Pharmacology, , New York University Langone Health, ; New York, NY USA
                [5 ]GRID grid.137628.9, ISNI 0000 0004 1936 8753, NYU Neuroscience Institute, , New York University Langone Health, ; New York, NY USA
                [6 ]GRID grid.10392.39, ISNI 0000 0001 2190 1447, Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, , University of Tübingen, ; Tübingen, Germany
                [7 ]GRID grid.137628.9, ISNI 0000 0004 1936 8753, Department of Cell Biology, , New York University Langone Health, ; New York, NY USA
                Author information
                http://orcid.org/0000-0002-0280-8375
                http://orcid.org/0000-0001-9259-3015
                http://orcid.org/0000-0002-7839-8878
                http://orcid.org/0000-0003-0138-7529
                http://orcid.org/0000-0001-6890-6763
                http://orcid.org/0000-0001-5124-1021
                Article
                1084
                10.1038/s41593-022-01084-8
                9174056
                35654956
                77de1bb6-eeb4-4e2e-a497-58da62e666e1
                © The Author(s) 2022

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 9 August 2021
                : 25 April 2022
                Funding
                Funded by: NIH P01AG017617
                Categories
                Article
                Custom metadata
                © The Author(s), under exclusive licence to Springer Nature America, Inc. 2022

                Neurosciences
                alzheimer's disease,autophagy,lysosomes,cellular neuroscience
                Neurosciences
                alzheimer's disease, autophagy, lysosomes, cellular neuroscience

                Comments

                Comment on this article