LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 244

Search options

  1. Article ; Online: Understanding the endo-lysosomal network in neurodegeneration.

    Cullen, Peter J / Holstege, Henne / Small, Scott A / St George-Hyslop, Peter

    Philosophical transactions of the Royal Society of London. Series B, Biological sciences

    2024  Volume 379, Issue 1899, Page(s) 20220372

    MeSH term(s) Humans ; Lysosomes ; Alzheimer Disease
    Language English
    Publishing date 2024-02-19
    Publishing country England
    Document type Journal Article
    ZDB-ID 208382-6
    ISSN 1471-2970 ; 0080-4622 ; 0264-3839 ; 0962-8436
    ISSN (online) 1471-2970
    ISSN 0080-4622 ; 0264-3839 ; 0962-8436
    DOI 10.1098/rstb.2022.0372
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article: Does Soluble TREM2 Protect Against Alzheimer's Disease?

    Brown, Guy C / St George-Hyslop, Peter

    Frontiers in aging neuroscience

    2022  Volume 13, Page(s) 834697

    Abstract: Triggering Receptor Expressed in Myeloid Cells 2 (TREM2) is a pattern recognition receptor on myeloid cells, and is upregulated on microglia surrounding amyloid plaques in Alzheimer's disease (AD). Rare, heterozygous mutations in TREM2 (e.g., R47H) ... ...

    Abstract Triggering Receptor Expressed in Myeloid Cells 2 (TREM2) is a pattern recognition receptor on myeloid cells, and is upregulated on microglia surrounding amyloid plaques in Alzheimer's disease (AD). Rare, heterozygous mutations in TREM2 (e.g., R47H) increase AD risk several fold. TREM2 can be cleaved at the plasma membrane by metalloproteases to release the ectodomain as soluble TREM2 (sTREM2). Wild-type sTREM2 binds oligomeric amyloid beta (Aβ) and acts as an extracellular chaperone, blocking and reversing Aβ oligomerization and fibrillization, and preventing Aβ-induced neuronal loss
    Language English
    Publishing date 2022-01-28
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2558898-9
    ISSN 1663-4365
    ISSN 1663-4365
    DOI 10.3389/fnagi.2021.834697
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Mimicking hypomethylation of FUS requires liquid-liquid phase separation to induce synaptic dysfunctions.

    Kim, Seung Chan / Mitchell, Scott J / Qamar, Seema / Whitcomb, Daniel J / Ruepp, Marc-David / St George-Hyslop, Peter / Cho, Kwangwook

    Acta neuropathologica communications

    2023  Volume 11, Issue 1, Page(s) 199

    Abstract: The hypomethylation of fused in sarcoma (FUS) in frontotemporal lobar degeneration promotes the formation of irreversible condensates of FUS. However, the mechanisms by which these hypomethylated FUS condensates cause neuronal dysfunction are unknown. ... ...

    Abstract The hypomethylation of fused in sarcoma (FUS) in frontotemporal lobar degeneration promotes the formation of irreversible condensates of FUS. However, the mechanisms by which these hypomethylated FUS condensates cause neuronal dysfunction are unknown. Here we report that expression of FUS constructs mimicking hypomethylated FUS causes aberrant dendritic FUS condensates in CA1 neurons. These hypomethylated FUS condensates exhibit spontaneous, and activity induced movement within the dendrite. They impair excitatory synaptic transmission, postsynaptic density-95 expression, and dendritic spine plasticity. These neurophysiological defects are dependent upon both the dendritic localisation of the condensates, and their ability to undergo liquid-liquid phase separation. These results indicate that the irreversible liquid-liquid phase separation is a key component of hypomethylated FUS pathophysiology in sporadic FTLD, and this can cause synapse dysfunction in sporadic FTLD.
    MeSH term(s) Humans ; Phase Separation ; RNA-Binding Protein FUS/genetics ; RNA-Binding Protein FUS/metabolism ; Frontotemporal Dementia ; Frontotemporal Lobar Degeneration/genetics ; DNA Methylation
    Chemical Substances RNA-Binding Protein FUS ; FUS protein, human
    Language English
    Publishing date 2023-12-18
    Publishing country England
    Document type Journal Article
    ZDB-ID 2715589-4
    ISSN 2051-5960 ; 2051-5960
    ISSN (online) 2051-5960
    ISSN 2051-5960
    DOI 10.1186/s40478-023-01703-w
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Spatially non-uniform condensates emerge from dynamically arrested phase separation.

    Erkamp, Nadia A / Sneideris, Tomas / Ausserwöger, Hannes / Qian, Daoyuan / Qamar, Seema / Nixon-Abell, Jonathon / St George-Hyslop, Peter / Schmit, Jeremy D / Weitz, David A / Knowles, Tuomas P J

    Nature communications

    2023  Volume 14, Issue 1, Page(s) 684

    Abstract: The formation of biomolecular condensates through phase separation from proteins and nucleic acids is emerging as a spatial organisational principle used broadly by living cells. Many such biomolecular condensates are not, however, homogeneous fluids, ... ...

    Abstract The formation of biomolecular condensates through phase separation from proteins and nucleic acids is emerging as a spatial organisational principle used broadly by living cells. Many such biomolecular condensates are not, however, homogeneous fluids, but possess an internal structure consisting of distinct sub-compartments with different compositions. Notably, condensates can contain compartments that are depleted in the biopolymers that make up the condensate. Here, we show that such double-emulsion condensates emerge via dynamically arrested phase transitions. The combination of a change in composition coupled with a slow response to this change can lead to the nucleation of biopolymer-poor droplets within the polymer-rich condensate phase. Our findings demonstrate that condensates with a complex internal architecture can arise from kinetic, rather than purely thermodynamic driving forces, and provide more generally an avenue to understand and control the internal structure of condensates in vitro and in vivo.
    MeSH term(s) Proteins ; Nucleic Acids ; Biopolymers ; Thermodynamics
    Chemical Substances Proteins ; Nucleic Acids ; Biopolymers
    Language English
    Publishing date 2023-02-08
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-023-36059-1
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Deciphering microglial diversity in Alzheimer's disease.

    Brown, Guy C / St George-Hyslop, Peter H

    Science (New York, N.Y.)

    2016  Volume 356, Issue 6343, Page(s) 1123–1124

    MeSH term(s) Alzheimer Disease ; Brain ; Humans ; Microglia
    Language English
    Publishing date 2016-11-30
    Publishing country United States
    Document type Journal Article ; Comment
    ZDB-ID 128410-1
    ISSN 1095-9203 ; 0036-8075
    ISSN (online) 1095-9203
    ISSN 0036-8075
    DOI 10.1126/science.aan7893
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: The liquid-to-solid transition of FUS is promoted by the condensate surface.

    Shen, Yi / Chen, Anqi / Wang, Wenyun / Shen, Yinan / Ruggeri, Francesco Simone / Aime, Stefano / Wang, Zizhao / Qamar, Seema / Espinosa, Jorge R / Garaizar, Adiran / St George-Hyslop, Peter / Collepardo-Guevara, Rosana / Weitz, David A / Vigolo, Daniele / Knowles, Tuomas P J

    Proceedings of the National Academy of Sciences of the United States of America

    2023  Volume 120, Issue 33, Page(s) e2301366120

    Abstract: A wide range of macromolecules can undergo phase separation, forming biomolecular condensates in living cells. These membraneless organelles are typically highly dynamic, formed reversibly, and carry out essential functions in biological systems. ... ...

    Abstract A wide range of macromolecules can undergo phase separation, forming biomolecular condensates in living cells. These membraneless organelles are typically highly dynamic, formed reversibly, and carry out essential functions in biological systems. Crucially, however, a further liquid-to-solid transition of the condensates can lead to irreversible pathological aggregation and cellular dysfunction associated with the onset and development of neurodegenerative diseases. Despite the importance of this liquid-to-solid transition of proteins, the mechanism by which it is initiated in normally functional condensates is unknown. Here we show, by measuring the changes in structure, dynamics, and mechanics in time and space, that single-component FUS condensates do not uniformly convert to a solid gel, but rather that liquid and gel phases coexist simultaneously within the same condensate, resulting in highly inhomogeneous structures. Furthermore, our results show that this transition originates at the interface between the condensate and the dilute continuous phase, and once initiated, the gelation process propagates toward the center of the condensate. To probe such spatially inhomogeneous rheology during condensate aging, we use a combination of established micropipette aspiration experiments together with two optical techniques, spatial dynamic mapping and reflective confocal dynamic speckle microscopy. These results reveal the importance of the spatiotemporal dimension of the liquid-to-solid transition and highlight the interface of biomolecular condensates as a critical element in driving pathological protein aggregation.
    MeSH term(s) Humans ; Biomolecular Condensates ; Microscopy, Confocal ; Protein Aggregation, Pathological ; Rheology ; RNA-Binding Protein FUS
    Chemical Substances FUS protein, human ; RNA-Binding Protein FUS
    Language English
    Publishing date 2023-08-07
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 209104-5
    ISSN 1091-6490 ; 0027-8424
    ISSN (online) 1091-6490
    ISSN 0027-8424
    DOI 10.1073/pnas.2301366120
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: MicroRNA-128 suppresses tau phosphorylation and reduces amyloid-beta accumulation by inhibiting the expression of GSK3β, APPBP2, and mTOR in Alzheimer's disease.

    Li, Siwen / Poon, Chi Him / Zhang, Zhigang / Yue, Ming / Chen, Ruijun / Zhang, Yalun / Hossain, Md Farhad / Pan, Yining / Zhao, Jun / Rong, Lei / Chu, Leung Wing / Shea, Yat Fung / Rogaeva, Ekaterina / Tu, Jie / St George-Hyslop, Peter / Lim, Lee Wei / Song, You-Qiang

    CNS neuroscience & therapeutics

    2023  Volume 29, Issue 7, Page(s) 1848–1864

    Abstract: Introduction and aims: Alzheimer's disease (AD) is characterized by the abnormal accumulation of hyperphosphorylated tau proteins and amyloid-beta (Aβ) peptides. Recent studies have shown that many microRNAs (miRNAs) are dysregulated in AD, and ... ...

    Abstract Introduction and aims: Alzheimer's disease (AD) is characterized by the abnormal accumulation of hyperphosphorylated tau proteins and amyloid-beta (Aβ) peptides. Recent studies have shown that many microRNAs (miRNAs) are dysregulated in AD, and modulation of these miRNAs can influence the development of tau and Aβ pathology. The brain-specific miRNA miR-128, encoded by MIR128-1 and MIR128-2, is important for brain development and dysregulated in AD. In this study, the role of miR-128 in tau and Aβ pathology as well as the regulatory mechanism underlying its dysregulation were investigated.
    Methods: The effect of miR-128 on tau phosphorylation and Aβ accumulation was examined in AD cellular models through miR-128 overexpression and inhibition. The therapeutic potential of miR-128 in AD mouse model was assessed by comparing phenotypes of 5XFAD mice administered with miR-128-expressing AAVs with 5XFAD mice administered with control AAVs. Phenotypes examined included behavior, plaque load, and protein expression. The regulatory factor of miR-128 transcription was identified through luciferase reporter assay and validated by siRNA knockdown and ChIP analysis.
    Results: Both gain-of-function and loss-of-function studies in AD cellular models reveal that miR-128 represses tau phosphorylation and Aβ secretion. Subsequent investigations show that miR-128 directly inhibits the expression of tau phosphorylation kinase GSK3β and Aβ modulators APPBP2 and mTOR. Upregulation of miR-128 in the hippocampus of 5XFAD mice ameliorates learning and memory impairments, decreases plaque deposition, and enhances autophagic flux. We further demonstrated that C/EBPα transactivates MIR128-1 transcription, while both C/EBPα and miR-128 expression are inhibited by Aβ.
    Conclusion: Our findings suggest that miR-128 suppresses AD pathogenesis, and could be a promising therapeutic target for AD. We also find a possible mechanism underlying the dysregulation of miR-128 in AD, in which Aβ reduces miR-128 expression by inhibiting C/EBPα.
    MeSH term(s) Mice ; Animals ; Alzheimer Disease/metabolism ; MicroRNAs/metabolism ; Phosphorylation ; Glycogen Synthase Kinase 3 beta ; Mice, Transgenic ; Amyloid beta-Peptides/metabolism ; tau Proteins/metabolism ; Disease Models, Animal ; TOR Serine-Threonine Kinases/metabolism
    Chemical Substances MicroRNAs ; Glycogen Synthase Kinase 3 beta (EC 2.7.11.1) ; Amyloid beta-Peptides ; tau Proteins ; TOR Serine-Threonine Kinases (EC 2.7.11.1) ; Mirn128 microRNA, mouse
    Language English
    Publishing date 2023-03-07
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2423461-8
    ISSN 1755-5949 ; 1755-5930
    ISSN (online) 1755-5949
    ISSN 1755-5930
    DOI 10.1111/cns.14143
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Regulation of membrane fluidity by RNF145-triggered degradation of the lipid hydrolase ADIPOR2.

    Volkmar, Norbert / Gawden-Bone, Christian M / Williamson, James C / Nixon-Abell, Jonathon / West, James A / St George-Hyslop, Peter H / Kaser, Arthur / Lehner, Paul J

    The EMBO journal

    2022  Volume 41, Issue 19, Page(s) e110777

    Abstract: The regulation of membrane lipid composition is critical for cellular homeostasis. Cells are particularly sensitive to phospholipid saturation, with increased saturation causing membrane rigidification and lipotoxicity. How mammalian cells sense membrane ...

    Abstract The regulation of membrane lipid composition is critical for cellular homeostasis. Cells are particularly sensitive to phospholipid saturation, with increased saturation causing membrane rigidification and lipotoxicity. How mammalian cells sense membrane lipid composition and reverse fatty acid (FA)-induced membrane rigidification is poorly understood. Here we systematically identify proteins that differ between mammalian cells fed saturated versus unsaturated FAs. The most differentially expressed proteins were two ER-resident polytopic membrane proteins: the E3 ubiquitin ligase RNF145 and the lipid hydrolase ADIPOR2. In unsaturated lipid membranes, RNF145 is stable, promoting its lipid-sensitive interaction, ubiquitination and degradation of ADIPOR2. When membranes become enriched in saturated FAs, RNF145 is rapidly auto-ubiquitinated and degraded, stabilising ADIPOR2, whose hydrolase activity restores lipid homeostasis and prevents lipotoxicity. We therefore identify RNF145 as a FA-responsive ubiquitin ligase which, together with ADIPOR2, defines an autoregulatory pathway that controls cellular membrane lipid homeostasis and prevents acute lipotoxic stress.
    MeSH term(s) Animals ; Fatty Acids/metabolism ; Hydrolases/metabolism ; Mammals ; Membrane Fluidity ; Membrane Proteins/metabolism ; Phospholipids ; Ubiquitin/metabolism ; Ubiquitin-Protein Ligases/genetics ; Ubiquitin-Protein Ligases/metabolism
    Chemical Substances Fatty Acids ; Membrane Proteins ; Phospholipids ; Ubiquitin ; Ubiquitin-Protein Ligases (EC 2.3.2.27) ; Hydrolases (EC 3.-)
    Language English
    Publishing date 2022-08-22
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 586044-1
    ISSN 1460-2075 ; 0261-4189
    ISSN (online) 1460-2075
    ISSN 0261-4189
    DOI 10.15252/embj.2022110777
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Withdrawal: Cytosolic proteins regulate α-synuclein dissociation from presynaptic membranes.

    Wislet-Gendebien, Sabine / D'Souza, Cheryl / Kawarai, Toshitaka / St George-Hyslop, Peter / Westaway, David / Fraser, Paul / Tandon, Anurag

    The Journal of biological chemistry

    2020  Volume 295, Issue 39, Page(s) 13694

    Language English
    Publishing date 2020-12-24
    Publishing country United States
    Document type Journal Article ; Retraction of Publication
    ZDB-ID 2997-x
    ISSN 1083-351X ; 0021-9258
    ISSN (online) 1083-351X
    ISSN 0021-9258
    DOI 10.1074/jbc.W120.015777
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Structural and Chemical Biology of Presenilin Complexes.

    Johnson, Douglas S / Li, Yue-Ming / Pettersson, Martin / St George-Hyslop, Peter H

    Cold Spring Harbor perspectives in medicine

    2017  Volume 7, Issue 12

    Abstract: The presenilin proteins are the catalytic subunits of a tetrameric complex containing presenilin 1 or 2, anterior pharynx defective 1 (APH1), nicastrin, and PEN-2. Other components such as TMP21 may exist in a subset of specialized complexes. The ... ...

    Abstract The presenilin proteins are the catalytic subunits of a tetrameric complex containing presenilin 1 or 2, anterior pharynx defective 1 (APH1), nicastrin, and PEN-2. Other components such as TMP21 may exist in a subset of specialized complexes. The presenilin complex is the founding member of a unique class of aspartyl proteases that catalyze the γ, ɛ, ζ site cleavage of the transmembrane domains of Type I membrane proteins including amyloid precursor protein (APP) and Notch. Here, we detail the structural and chemical biology of this unusual enzyme. Taken together, these studies suggest that the complex exists in several conformations, and subtle long-range (allosteric) shifts in the conformation of the complex underpin substrate access to the catalytic site and the mechanism of action for allosteric inhibitors and modulators. Understanding the mechanics of these shifts will facilitate the design of γ-secretase modulator (GSM) compounds that modulate the relative efficiency of γ, ɛ, ζ site cleavage and/or substrate specificity.
    MeSH term(s) Amyloid beta-Protein Precursor/chemistry ; Amyloid beta-Protein Precursor/metabolism ; Animals ; Humans ; Presenilin-1/chemistry ; Presenilin-1/metabolism ; Presenilin-2/chemistry ; Presenilin-2/metabolism
    Chemical Substances Amyloid beta-Protein Precursor ; Presenilin-1 ; Presenilin-2
    Language English
    Publishing date 2017-12-01
    Publishing country United States
    Document type Journal Article ; Review
    ISSN 2157-1422
    ISSN (online) 2157-1422
    DOI 10.1101/cshperspect.a024067
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top