1
|
Schultz SA, Liu L, Schultz AP, Fitzpatrick CD, Levin R, Bellier JP, Shirzadi Z, Joseph-Mathurin N, Chen CD, Benzinger TLS, Day GS, Farlow MR, Gordon BA, Hassenstab JJ, Jack CR, Jucker M, Karch CM, Lee JH, Levin J, Perrin RJ, Schofield PR, Xiong C, Johnson KA, McDade E, Bateman RJ, Sperling RA, Selkoe DJ, Chhatwal JP. γ-Secretase activity, clinical features, and biomarkers of autosomal dominant Alzheimer's disease: cross-sectional and longitudinal analysis of the Dominantly Inherited Alzheimer Network observational study (DIAN-OBS). Lancet Neurol 2024; 23:913-924. [PMID: 39074479 DOI: 10.1016/s1474-4422(24)00236-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/26/2024] [Accepted: 05/28/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Genetic variants that cause autosomal dominant Alzheimer's disease are highly penetrant but vary substantially regarding age at symptom onset (AAO), rates of cognitive decline, and biomarker changes. Most pathogenic variants that cause autosomal dominant Alzheimer's disease are in presenilin 1 (PSEN1), which encodes the catalytic core of γ-secretase, an enzyme complex that is crucial in production of amyloid β. We aimed to investigate whether the heterogeneity in AAO and biomarker trajectories in carriers of PSEN1 pathogenic variants could be predicted on the basis of the effects of individual PSEN1 variants on γ-secretase activity and amyloid β production. METHODS For this cross-sectional and longitudinal analysis, we used data from participants enrolled in the Dominantly Inherited Alzheimer Network observational study (DIAN-OBS) via the DIAN-OBS data freeze version 15 (data collected between Feb 29, 2008, and June 30, 2020). The data freeze included data from 20 study sites in research institutions, universities, hospitals, and clinics across Europe, North and South America, Asia, and Oceania. We included individuals with PSEN1 pathogenic variants for whom relevant genetic, clinical, imaging, and CSF data were available. PSEN1 pathogenic variants were characterised via genetically modified PSEN1 and PSEN2 double-knockout human embryonic kidney 293T cells and immunoassays for Aβ37, Aβ38, Aβ40, Aβ42, and Aβ43. A summary measure of γ-secretase activity (γ-secretase composite [GSC]) was calculated for each variant and compared with clinical history-derived AAO using correlation analyses. We used linear mixed-effect models to assess associations between GSC scores and multimodal-biomarker and clinical data from DIAN-OBS. We used separate models to assess associations with Clinical Dementia Rating Sum of Boxes (CDR-SB), Mini-Mental State Examination (MMSE), and Wechsler Memory Scale-Revised (WMS-R) Logical Memory Delayed Recall, [11C]Pittsburgh compound B (PiB)-PET and brain glucose metabolism using [18F] fluorodeoxyglucose (FDG)-PET, CSF Aβ42-to-Aβ40 ratio (Aβ42/40), CSF log10 (phosphorylated tau 181), CSF log10 (phosphorylated tau 217), and MRI-based hippocampal volume. FINDINGS Data were included from 190 people carrying PSEN1 pathogenic variants, among whom median age was 39·0 years (IQR 32·0 to 48·0) and AAO was 44·5 years (40·6 to 51·4). 109 (57%) of 190 carriers were female and 81 (43%) were male. Lower GSC values (ie, lower γ-secretase activity than wild-type PSEN1) were associated with earlier AAO (r=0·58; p<0·0001). GSC was associated with MMSE (β=0·08, SE 0·03; p=0·0043), CDR-SB (-0·05, 0·02; p=0·0027), and WMS-R Logical Memory Delayed Recall scores (0·09, 0·02; p=0·0006). Lower GSC values were associated with faster increase in PiB-PET signal (p=0·0054), more rapid decreases in hippocampal volume (4·19, 0·77; p<0·0001), MMSE (0·02, 0·01; p=0·0020), and WMS-R Logical Memory Delayed Recall (0·004, 0·001; p=0·0003). INTERPRETATION Our findings suggest that clinical heterogeneity in people with autosomal dominant Alzheimer's disease can be at least partly explained by different effects of PSEN1 variants on γ-secretase activity and amyloid β production. They support targeting γ-secretase as a therapeutic approach and suggest that cell-based models could be used to improve prediction of symptom onset. FUNDING US National Institute on Aging, Alzheimer's Association, German Center for Neurodegenerative Diseases, Raul Carrea Institute for Neurological Research, Japan Agency for Medical Research and Development, Korea Health Industry Development Institute, South Korean Ministry of Health and Welfare, South Korean Ministry of Science and ICT, and Spanish Institute of Health Carlos III.
Collapse
Affiliation(s)
- Stephanie A Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Medical School, Harvard University, Boston, MA, USA
| | - Lei Liu
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Boston, MA, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Medical School, Harvard University, Boston, MA, USA
| | - Colleen D Fitzpatrick
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Medical School, Harvard University, Boston, MA, USA
| | - Raina Levin
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Medical School, Harvard University, Boston, MA, USA
| | - Jean-Pierre Bellier
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Boston, MA, USA
| | - Zahra Shirzadi
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Medical School, Harvard University, Boston, MA, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Charles D Chen
- Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, USA
| | | | - Gregory S Day
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Martin R Farlow
- Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, USA
| | | | | | - Mathias Jucker
- German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Celeste M Karch
- Department of Psychiatry, Washington University, St Louis, MO, USA
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Johannes Levin
- German Center for Neurodegenerative Diseases, Munich, Germany; Department of Neurology, Ludwig Maximilian University of Munich, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany
| | - Richard J Perrin
- Department of Psychiatry, Washington University, St Louis, MO, USA; Department of Pathology and Immunology, Washington University, St Louis, MO, USA
| | - Peter R Schofield
- Neuroscience Research Australia, Randwick, NSW, Australia; School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Chengjie Xiong
- Division of Biostatistics, Washington University, St Louis, MO, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Medical School, Harvard University, Boston, MA, USA
| | - Eric McDade
- Department of Neurology, Washington University, St Louis, MO, USA
| | | | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Medical School, Harvard University, Boston, MA, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Dennis J Selkoe
- Department of Neurology, Medical School, Harvard University, Boston, MA, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Boston, MA, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Medical School, Harvard University, Boston, MA, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Boston, MA, USA.
| |
Collapse
|
2
|
Wenzel TJ, Desjarlais JD, Mousseau DD. Human brain organoids containing microglia that have arisen innately adapt to a β-amyloid challenge better than those in which microglia are integrated by co-culture. Stem Cell Res Ther 2024; 15:258. [PMID: 39135132 PMCID: PMC11320858 DOI: 10.1186/s13287-024-03876-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Alzheimer disease (AD) is a heterogenous and multifactorial disease, and its pathology is partly driven by microglia and their activated phenotype. Brain organoids (BOs) are gaining prominence as a relevant model of the human brain for the study of AD; however, BOs are commonly devoid of microglia. To overcome this limitation, current protocols incorporate microglia through either (1) co-culture (BO co-culture), or (2) molecular manipulation at critical windows of BO development to have microglia arise innately (BO innate cultures). It is currently unclear whether the microglia incorporated into BOs by either of these two protocols differ in function. METHODS At in vitro day 90, BO innate cultures and BO-co-cultures were challenged with the AD-related β-amyloid peptide (Aβ) for up to 72 h. After Aβ challenge, BOs were collected for immunoblotting. Immunoblots compared immunodensity and protein banding of Aβ and ionized calcium-binding adapter molecule 1 (IBA1, a marker of microglial activation) in BOs. The translational potential of these observations was supported using 56 human cortical samples from neurocognitively normal donors and patients with early-onset AD and late-onset AD. Statistical analyses were conducted using the Kruskal-Wallis test, a two-way ANOVA, or a simple linear regression, and where applicable, followed by Dunn's or Sidak's test. RESULTS We show that BO co-cultures promote Aβ oligomerization as early as 24 h and this coincides with a significant increase in IBA1 levels. In contrast, the Aβs do not oligomerize in BO innate cultures and the IBA1 response was modest and only emerged after 48 h. In human cortical samples, we found IBA1 levels correlated with age at onset, age at death, and the putative diagnostic Aβ(1-42)/Aβ(1-40) ratio (particularly in their oligomeric forms) in a sex-dependent manner. CONCLUSIONS Our unique observations suggest that BOs with innate microglia model the response of a healthy brain to Aβ, and by extension the initial stages of Aβ challenge. It would be impossible to model these early stages of pathogenesis in BOs where microglia are already compromised, such as those with microglia incorporated by co-culture.
Collapse
Affiliation(s)
- Tyler J Wenzel
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
| | - Joseph D Desjarlais
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| |
Collapse
|
3
|
De Strooper B, Karran E. New precision medicine avenues to the prevention of Alzheimer's disease from insights into the structure and function of γ-secretases. EMBO J 2024; 43:887-903. [PMID: 38396302 PMCID: PMC10943082 DOI: 10.1038/s44318-024-00057-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: 11/18/2023] [Revised: 01/20/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Two phase-III clinical trials with anti-amyloid peptide antibodies have met their primary goal, i.e. slowing of Alzheimer's disease (AD) progression. However, antibody therapy may not be the optimal therapeutic modality for AD prevention, as we will discuss in the context of the earlier small molecules described as "γ-secretase modulators" (GSM). We review here the structure, function, and pathobiology of γ-secretases, with a focus on how mutations in presenilin genes result in early-onset AD. Significant progress has been made in generating compounds that act in a manner opposite to pathogenic presenilin mutations: they stabilize the proteinase-substrate complex, thereby increasing the processivity of substrate cleavage and altering the size spectrum of Aβ peptides produced. We propose the term "γ-secretase allosteric stabilizers" (GSAS) to distinguish these compounds from the rather heterogenous class of GSM. The GSAS represent, in theory, a precision medicine approach to the prevention of amyloid deposition, as they specifically target a discrete aspect in a complex cell biological signalling mechanism that initiates the pathological processes leading to Alzheimer's disease.
Collapse
Affiliation(s)
- Bart De Strooper
- Dementia Research Institute, Institute of Neurology, University College London, at the Francis Crick Institute, London, NW1 AT, UK.
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain & Disease Research, and Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium.
| | - Eric Karran
- Cambridge Research Center, AbbVie, Inc., Cambridge, MA, USA
| |
Collapse
|
4
|
Cournut A, Moustiez P, Coffinier Y, Enjalbal C, Bich C. Innovative SALDI mass spectrometry analysis for Alzheimer's disease synthetic peptides detection. Talanta 2024; 268:125357. [PMID: 37951181 DOI: 10.1016/j.talanta.2023.125357] [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: 07/04/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/13/2023]
Abstract
Alzheimer's disease (AD) is nowadays the prominent cause of senile dementia. This pathology is characterized by aggregation of neurofibrillary tangles in cells and by the accumulation of amyloid plaques in the brain. Noteworthy, a phosphorylated protein (tau protein) and a peptide presenting two overlapping sequences of 40 or 42 residues named β-amyloid peptides 1-40 (Aβ 1-40) and 1-42 (Aβ 1-42), respectively, were related to such deleterious phenomena. Singularly, the neurotoxicity was primarily attributed to the amyloid peptide Aβ 1-42 form due to its capacity to fold into beta-sheets rendering it insoluble thus causing subsequent aggregation and accumulation in vivo. Regarding AD diagnosis relying on mass spectrometry, Aβ 1-42 and/or Aβ 1-40 were considered as relevant biomarkers being measured in cerebrospinal fluids (CSF), blood and urine. Under that context, we aimed at implementing an innovative method to evidence the depletion of circulating Aβ 1-42 amyloid peptide compared to the shorter Aβ 1-40 form indicating a pathologic state. We investigated Surface-Assisted Laser Desorption/Ionization Mass Spectrometry (SALDI-MS) in order to monitor the Aβ 1-42/Aβ 1-40 ratio without any prior sample treatment or enrichment. Taking into account that β-amyloid peptide and 1-42 can aggregate into beta-sheets depending on the experimental conditions, specific attention was devoted to sample integrity monitoring performed by circular dichroism experiments during SALDI-MS method development.
Collapse
Affiliation(s)
- Aline Cournut
- Univ Montpellier, IBMM, CNRS, ENSCM, Montpellier, France
| | - Paul Moustiez
- Univ Lille, IEMN, UMR CNRS 8520, Villeneuve d'Ascq, France
| | | | | | - Claudia Bich
- Univ Montpellier, IBMM, CNRS, ENSCM, Montpellier, France.
| |
Collapse
|
5
|
Kepp KP, Robakis NK, Høilund-Carlsen PF, Sensi SL, Vissel B. The amyloid cascade hypothesis: an updated critical review. Brain 2023; 146:3969-3990. [PMID: 37183523 DOI: 10.1093/brain/awad159] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/16/2023] Open
Abstract
Results from recent clinical trials of antibodies that target amyloid-β (Aβ) for Alzheimer's disease have created excitement and have been heralded as corroboration of the amyloid cascade hypothesis. However, while Aβ may contribute to disease, genetic, clinical, imaging and biochemical data suggest a more complex aetiology. Here we review the history and weaknesses of the amyloid cascade hypothesis in view of the new evidence obtained from clinical trials of anti-amyloid antibodies. These trials indicate that the treatments have either no or uncertain clinical effect on cognition. Despite the importance of amyloid in the definition of Alzheimer's disease, we argue that the data point to Aβ playing a minor aetiological role. We also discuss data suggesting that the concerted activity of many pathogenic factors contribute to Alzheimer's disease and propose that evolving multi-factor disease models will better underpin the search for more effective strategies to treat the disease.
Collapse
Affiliation(s)
- Kasper P Kepp
- Section of Biophysical and Biomedicinal chemistry, DTU Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Nikolaos K Robakis
- Icahn School of Medicine at Mount Sinai Medical Center, New York, NY 10029, USA
| | - Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Stefano L Sensi
- Center for Advanced Studies and Technology-CAST, and Institute for Advanced Biotechnology (ITAB), University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
| | - Bryce Vissel
- St Vincent's Hospital Centre for Applied Medical Research, St Vincent's Hospital, Sydney, 2010, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, Sydney, NSW 2052, Australia
| |
Collapse
|
6
|
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: 0] [Impact Index Per Article: 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
|
7
|
Dawkins E, Derks RJE, Schifferer M, Trambauer J, Winkler E, Simons M, Paquet D, Giera M, Kamp F, Steiner H. Membrane lipid remodeling modulates γ-secretase processivity. J Biol Chem 2023; 299:103027. [PMID: 36805335 PMCID: PMC10070668 DOI: 10.1016/j.jbc.2023.103027] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/18/2023] Open
Abstract
Imbalances in the amounts of amyloid-β peptides (Aβ) generated by the membrane proteases β- and γ-secretase are considered as a trigger of Alzheimer´s disease (AD). Cell-free studies of γ-secretase have shown that increasing membrane thickness modulates Aβ generation, but it has remained unclear if these effects are translatable to cells. Here we show that the very long chain fatty acid erucic acid (EA) triggers acyl chain remodeling in AD cell models, resulting in substantial lipidome alterations which included increased esterification of EA in membrane lipids. Membrane remodeling enhanced γ-secretase processivity, resulting in the increased production of the potentially beneficial Aβ37 and/or Aβ38 species in multiple cell lines. Unexpectedly, we found that the membrane remodeling stimulated total Aβ secretion by cells expressing WT γ-secretase, but lowered it for cells expressing an aggressive familial AD mutant γ-secretase. We conclude that EA-mediated modulation of membrane composition is accompanied by complex lipid homeostatic changes that can impact amyloidogenic processing in different ways and elicit distinct γ-secretase responses, providing critical implications for lipid-based AD treatment strategies.
Collapse
Affiliation(s)
- Edgar Dawkins
- Division of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU Munich, Munich, Germany
| | - Rico J E Derks
- Center for Proteomics & Metabolomics, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Johannes Trambauer
- Division of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU Munich, Munich, Germany
| | - Edith Winkler
- Division of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU Munich, Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany; Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Dominik Paquet
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Martin Giera
- Center for Proteomics & Metabolomics, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Frits Kamp
- Division of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU Munich, Munich, Germany
| | - Harald Steiner
- Division of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
8
|
Petit D, Fernández SG, Zoltowska KM, Enzlein T, Ryan NS, O'Connor A, Szaruga M, Hill E, Vandenberghe R, Fox NC, Chávez-Gutiérrez L. Aβ profiles generated by Alzheimer's disease causing PSEN1 variants determine the pathogenicity of the mutation and predict age at disease onset. Mol Psychiatry 2022; 27:2821-2832. [PMID: 35365805 PMCID: PMC9156411 DOI: 10.1038/s41380-022-01518-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 02/05/2023]
Abstract
Familial Alzheimer's disease (FAD), caused by mutations in Presenilin (PSEN1/2) and Amyloid Precursor Protein (APP) genes, is associated with an early age at onset (AAO) of symptoms. AAO is relatively consistent within families and between carriers of the same mutations, but differs markedly between individuals carrying different mutations. Gaining a mechanistic understanding of why certain mutations manifest several decades earlier than others is extremely important in elucidating the foundations of pathogenesis and AAO. Pathogenic mutations affect the protease (PSEN/γ-secretase) and the substrate (APP) that generate amyloid β (Aβ) peptides. Altered Aβ metabolism has long been associated with AD pathogenesis, with absolute or relative increases in Aβ42 levels most commonly implicated in the disease development. However, analyses addressing the relationships between these Aβ42 increments and AAO are inconsistent. Here, we investigated this central aspect of AD pathophysiology via comprehensive analysis of 25 FAD-linked Aβ profiles. Hypothesis- and data-driven approaches demonstrate linear correlations between mutation-driven alterations in Aβ profiles and AAO. In addition, our studies show that the Aβ (37 + 38 + 40) / (42 + 43) ratio offers predictive value in the assessment of 'unclear' PSEN1 variants. Of note, the analysis of PSEN1 variants presenting additionally with spastic paraparesis, indicates that a different mechanism underlies the aetiology of this distinct clinical phenotype. This study thus delivers valuable assays for fundamental, clinical and genetic research as well as supports therapeutic interventions aimed at shifting Aβ profiles towards shorter Aβ peptides.
Collapse
Affiliation(s)
- Dieter Petit
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Sara Gutiérrez Fernández
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Katarzyna Marta Zoltowska
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Thomas Enzlein
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack Str. 10, 68163, Mannheim, Germany
| | - Natalie S Ryan
- UK Dementia Research Institute at UCL, Queen Square, WC1N 3BG, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Antoinette O'Connor
- UK Dementia Research Institute at UCL, Queen Square, WC1N 3BG, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Maria Szaruga
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Elizabeth Hill
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Herestraat 49 box 1027, 3000, Leuven, Belgium
- Neurology Department, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Nick C Fox
- UK Dementia Research Institute at UCL, Queen Square, WC1N 3BG, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Lucía Chávez-Gutiérrez
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium.
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium.
| |
Collapse
|
9
|
Potjewyd FM, Annor‐Gyamfi JK, Aubé J, Chu S, Conlon IL, Frankowski KJ, Guduru SKR, Hardy BP, Hopkins MD, Kinoshita C, Kireev DB, Mason ER, Moerk CT, Nwogbo F, Pearce KH, Richardson TI, Rogers DA, Soni DM, Stashko M, Wang X, Wells C, Willson TM, Frye SV, Young JE, Axtman AD. Use of AD Informer Set compounds to explore validity of novel targets in Alzheimer's disease pathology. ALZHEIMER'S & DEMENTIA: TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2022; 8:e12253. [PMID: 35434254 PMCID: PMC9005681 DOI: 10.1002/trc2.12253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/29/2021] [Accepted: 12/15/2021] [Indexed: 12/03/2022]
Abstract
Introduction A chemogenomic set of small molecules with annotated activities and implicated roles in Alzheimer's disease (AD) called the AD Informer Set was recently developed and made available to the AD research community: https://treatad.org/data‐tools/ad‐informer‐set/. Methods Small subsets of AD Informer Set compounds were selected for AD‐relevant profiling. Nine compounds targeting proteins expressed by six AD‐implicated genes prioritized for study by Target Enablement to Accelerate Therapy Development for Alzheimer's Disease (TREAT‐AD) teams were selected for G‐protein coupled receptor (GPCR), amyloid beta (Aβ) and tau, and pharmacokinetic (PK) studies. Four non‐overlapping compounds were analyzed in microglial cytotoxicity and phagocytosis assays. Results The nine compounds targeting CAPN2, EPHX2, MDK, MerTK/FLT3, or SYK proteins were profiled in 46 to 47 primary GPCR binding assays. Human induced pluripotent stem cell (iPSC)‐derived neurons were treated with the same nine compounds and secretion of Aβ peptides (Aβ40 and Aβ42) as well as levels of phosphophorylated tau (p‐tau, Thr231) and total tau (t‐tau) peptides measured at two concentrations and two timepoints. Finally, CD1 mice were dosed intravenously to determine preliminary PK and/or brain‐specific penetrance values for these compounds. As a final cell‐based study, a non‐overlapping subset of four compounds was selected based on single‐concentration screening for analysis of both cytotoxicity and phagocytosis in murine and human microglia cells. Discussion We have demonstrated the utility of the AD Informer Set in the validation of novel AD hypotheses using biochemical, cellular (primary and immortalized), and in vivo studies. The selectivity for their primary targets versus essential GPCRs in the brain was established for our compounds. Statistical changes in tau, p‐tau, Aβ40, and/or Aβ42 and blood–brain barrier penetrance were observed, solidifying the utility of specific compounds for AD. Single‐concentration phagocytosis results were validated as predictive of dose–response findings. These studies established workflows, validated assays, and illuminated next steps for protein targets and compounds.
Collapse
Affiliation(s)
- Frances M. Potjewyd
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Structural Genomics Consortium Chapel Hill North Carolina USA
| | - Joel K. Annor‐Gyamfi
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Structural Genomics Consortium Chapel Hill North Carolina USA
| | - Jeffrey Aubé
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Shaoyou Chu
- Department of Laboratory Medicine and Pathology University of Washington Seattle Washington USA
| | - Ivie L. Conlon
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Kevin J. Frankowski
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Shiva K. R. Guduru
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Brian P. Hardy
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Megan D. Hopkins
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Chizuru Kinoshita
- Department of Laboratory Medicine and Pathology University of Washington Seattle Washington USA
- Institute for Stem Cell and Regenerative Medicine University of Washington Seattle Washington USA
| | - Dmitri B. Kireev
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Emily R. Mason
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - Charles T. Moerk
- Department of Laboratory Medicine and Pathology University of Washington Seattle Washington USA
- Institute for Stem Cell and Regenerative Medicine University of Washington Seattle Washington USA
| | - Felix Nwogbo
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Kenneth H. Pearce
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Timothy I. Richardson
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - David A. Rogers
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Disha M. Soni
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - Michael Stashko
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Xiaodong Wang
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Carrow Wells
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Structural Genomics Consortium Chapel Hill North Carolina USA
| | - Timothy M. Willson
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Structural Genomics Consortium Chapel Hill North Carolina USA
| | - Stephen V. Frye
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Jessica E. Young
- Department of Laboratory Medicine and Pathology University of Washington Seattle Washington USA
- Institute for Stem Cell and Regenerative Medicine University of Washington Seattle Washington USA
| | - Alison D. Axtman
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Structural Genomics Consortium Chapel Hill North Carolina USA
| |
Collapse
|
10
|
Xu B, He Y, Liu L, Ye G, Chen L, Wang Q, Chen M, Chen Y, Long D. The Effects of Physical Running on Dendritic Spines and Amyloid-beta Pathology in 3xTg-AD Male Mice. Aging Dis 2022; 13:1293-1310. [PMID: 35855335 PMCID: PMC9286906 DOI: 10.14336/ad.2022.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/10/2022] [Indexed: 11/01/2022] Open
Abstract
Memory loss is the key symptom of Alzheimer's disease (AD). As successful drug treatments have not yet been identified, non-pharmaceutical interventions such as physical exercise and training have been employed to improve the memory function of people with dementia. We investigated the effect of prolonged physical running on hippocampal-dependent spatial memory and its underlying mechanisms using a well-established rodent model of AD. 3xTg-AD transgenic mice and non-transgenic mice were subjected to voluntary wheel running for 5 months (1 hour per day, 5 days per week), followed by spatial memory testing. After the behavioral testing, dendritic spines, synapses, and synaptic proteins as well as amyloid-beta (Aβ) pathology were analyzed in the dorsal hippocampi. Running improved hippocampal-dependent spatial memory in 3xTg-AD mice. This running strategy prevented both thin and mushroom-type spines on CA1 pyramidal cells in 3xTg-AD mice, whereas the effects of running in non-transgenic mice were limited to thin spines. The enormous effects of running on spines were accompanied by an increased number of synapses and upregulated expression of synaptic proteins. Notably, running downregulated the processing of amyloid precursor protein, decreasing intracellular APP expression and extracellular Aβ accumulation, and spatial memory performance correlated with levels of Aβ peptides Aβ1-40 and Aβ1-42. These data suggest that prolonged running may improve memory in preclinical AD via slowing down the amyloid pathology and preventing the loss of synaptic contacts.
Collapse
Affiliation(s)
- Benke Xu
- Department of Human Anatomy, School of Basic Medical Sciences, Yangtze University, Hubei 434023, China.
| | - Yun He
- Department of Human Anatomy, School of Basic Medical Sciences, Yangtze University, Hubei 434023, China.
| | - Lian Liu
- Department of Pharmacology, School of Basic Medical Sciences, Yangtze University, Hubei 434023, China.
| | - Guosheng Ye
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Lulu Chen
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Qingning Wang
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Michael Chen
- University of California, Los Angeles, CA 90095, USA.
| | - Yuncai Chen
- Department of Pediatrics, University of California, Irvine, CA 92697, USA.
- Correspondence should be addressed to: Dr. Dahong Long, Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China. E-mail: or Dr. Yuncai Chen, Department of Pediatrics, University of California-Irvine, Irvine, California 92697, USA. E-mail:
| | - Dahong Long
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
- Correspondence should be addressed to: Dr. Dahong Long, Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China. E-mail: or Dr. Yuncai Chen, Department of Pediatrics, University of California-Irvine, Irvine, California 92697, USA. E-mail:
| |
Collapse
|
11
|
Mehra R, Kepp KP. Computational prediction and molecular mechanism of γ-secretase modulators. Eur J Pharm Sci 2021; 157:105626. [DOI: 10.1016/j.ejps.2020.105626] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022]
|
12
|
Quartey MO, Nyarko JNK, Maley JM, Barnes JR, Bolanos MAC, Heistad RM, Knudsen KJ, Pennington PR, Buttigieg J, De Carvalho CE, Leary SC, Parsons MP, Mousseau DD. The Aβ(1-38) peptide is a negative regulator of the Aβ(1-42) peptide implicated in Alzheimer disease progression. Sci Rep 2021; 11:431. [PMID: 33432101 PMCID: PMC7801637 DOI: 10.1038/s41598-020-80164-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
The pool of β-Amyloid (Aβ) length variants detected in preclinical and clinical Alzheimer disease (AD) samples suggests a diversity of roles for Aβ peptides. We examined how a naturally occurring variant, e.g. Aβ(1-38), interacts with the AD-related variant, Aβ(1-42), and the predominant physiological variant, Aβ(1-40). Atomic force microscopy, Thioflavin T fluorescence, circular dichroism, dynamic light scattering, and surface plasmon resonance reveal that Aβ(1-38) interacts differently with Aβ(1-40) and Aβ(1-42) and, in general, Aβ(1-38) interferes with the conversion of Aβ(1-42) to a β-sheet-rich aggregate. Functionally, Aβ(1-38) reverses the negative impact of Aβ(1-42) on long-term potentiation in acute hippocampal slices and on membrane conductance in primary neurons, and mitigates an Aβ(1-42) phenotype in Caenorhabditis elegans. Aβ(1-38) also reverses any loss of MTT conversion induced by Aβ(1-40) and Aβ(1-42) in HT-22 hippocampal neurons and APOE ε4-positive human fibroblasts, although the combination of Aβ(1-38) and Aβ(1-42) inhibits MTT conversion in APOE ε4-negative fibroblasts. A greater ratio of soluble Aβ(1-42)/Aβ(1-38) [and Aβ(1-42)/Aβ(1-40)] in autopsied brain extracts correlates with an earlier age-at-death in males (but not females) with a diagnosis of AD. These results suggest that Aβ(1-38) is capable of physically counteracting, potentially in a sex-dependent manner, the neuropathological effects of the AD-relevant Aβ(1-42).
Collapse
Affiliation(s)
- Maa O Quartey
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada
| | - Jennifer N K Nyarko
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada
| | - Jason M Maley
- Saskatchewan Structural Sciences Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jocelyn R Barnes
- Division of BioMedical Sciences (Neurosciences), Memorial University of Newfoundland, St. John's, NL, Canada
| | | | - Ryan M Heistad
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada
| | - Kaeli J Knudsen
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada
| | - Paul R Pennington
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada
| | - Josef Buttigieg
- Department of Biology, University of Regina, Regina, SK, Canada
| | | | - Scot C Leary
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Matthew P Parsons
- Division of BioMedical Sciences (Neurosciences), Memorial University of Newfoundland, St. John's, NL, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
13
|
A computer-simulated mechanism of familial Alzheimer’s disease: Mutations enhance thermal dynamics and favor looser substrate-binding to γ-secretase. J Struct Biol 2020; 212:107648. [DOI: 10.1016/j.jsb.2020.107648] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/22/2020] [Accepted: 10/09/2020] [Indexed: 11/22/2022]
|
14
|
Qiu Q, Shen L, Jia L, Wang Q, Li F, Li Y, Jia J. A Novel PSEN1 M139L Mutation Found in a Chinese Pedigree with Early-Onset Alzheimer's Disease Increases Aβ42/Aβ40 ratio. J Alzheimers Dis 2020; 69:199-212. [PMID: 30958370 DOI: 10.3233/jad-181291] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Presenilin1 (PSEN1) is the most common gene related to familial Alzheimer's disease (AD). Only several mutation types from Chinese have been reported, with less biological function research conducted. OBJECTIVES We explore the pathological function of PSEN1 M139L, a mutation located at α-helix of PSEN1 transmembrane 2, using predictive programs and in vitro study and compare its effects on Aβ production to those of an artificial PSEN1 S141G located at non α-helix mutation face. METHODS APP, PSEN1, and PSEN2 genes were screened for mutations using Sanger sequencing in the DNA samples of the proband and additional available family members. Disease-mutation cosegregation analysis and three software programs were performed to predict the mutation's pathogenicity. In vitro, we investigated the impact of these mutations on Aβ production in HEK293-APPswe cells using lentiviral vectors harboring PSEN1 WT, PSEN1 M139L, the positive control (PSEN1 M139V) and the non α-helical mutation (PSEN1 S141G). In addition, we co-transfected PSEN1 and tau into cells to determine the mutations' impact on tau phosphorylation. RESULTS PSEN1 M139L mutation was discovered in the index patient and four affected siblings. Cosegregation analysis and silicon prediction suggested the mutation was probably disease causing. In vitro studies demonstrated that both PSEN1 M139L and PSEN1 S141G caused elevated ratios of Aβ42/Aβ40, but changes of tau phosphorylation were not detected. CONCLUSION The novel PSEN1 M139L mutation found in familial AD increases the Aβ42/Aβ40 ratio significantly. Mutations at non α-helical mutation face of PSEN1 TM2 can affect Aβ production and the region may play a key role in PSEN1 function.
Collapse
Affiliation(s)
- Qiongqiong Qiu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, P.R. Beijing, China
| | - Luxi Shen
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, P.R. Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, P.R. Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, P.R. Beijing, China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, P.R. Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.,National Clinical Research Center for Geriatric Disorders, Beijing, P.R. China
| | - Qi Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, P.R. Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, P.R. Beijing, China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, P.R. Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.,National Clinical Research Center for Geriatric Disorders, Beijing, P.R. China
| | - Fangyu Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, P.R. Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, P.R. Beijing, China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, P.R. Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.,National Clinical Research Center for Geriatric Disorders, Beijing, P.R. China
| | - Ying Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, P.R. Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, P.R. Beijing, China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, P.R. Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.,National Clinical Research Center for Geriatric Disorders, Beijing, P.R. China
| | - Jianping Jia
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, P.R. Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, P.R. Beijing, China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, P.R. Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.,National Clinical Research Center for Geriatric Disorders, Beijing, P.R. China
| |
Collapse
|
15
|
Hitzenberger M, Götz A, Menig S, Brunschweiger B, Zacharias M, Scharnagl C. The dynamics of γ-secretase and its substrates. Semin Cell Dev Biol 2020; 105:86-101. [DOI: 10.1016/j.semcdb.2020.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 12/18/2022]
|
16
|
Dehury B, Kepp KP. Membrane dynamics of γ-secretase with the anterior pharynx-defective 1B subunit. J Cell Biochem 2020; 122:69-85. [PMID: 32830360 DOI: 10.1002/jcb.29832] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/13/2020] [Indexed: 01/12/2023]
Abstract
The four-subunit protease complex γ-secretase cleaves many single-pass transmembrane (TM) substrates, including Notch and β-amyloid precursor protein to generate amyloid-β (Aβ), central to Alzheimer's disease. Two of the subunits anterior pharynx-defective 1 (APH-1) and presenilin (PS) exist in two homologous forms APH1-A and APH1-B, and PS1 and PS2. The consequences of these variations are poorly understood and could affect Aβ production and γ-secretase medicine. Here, we developed the first complete structural model of the APH-1B subunit using the published cryo-electron microscopy (cryo-EM) structures of APH1-A (Protein Data Bank: 5FN2, 5A63, and 6IYC). We then performed all-atom molecular dynamics simulations at 303 K in a realistic bilayer system to understand both APH-1B alone and in γ-secretase without and with substrate C83-bound. We show that APH-1B adopts a 7TM topology with a water channel topology similar to APH-1A. We demonstrate direct transport of water through this channel, mainly via Glu84, Arg87, His170, and His196. The apo and holo states closely resemble the experimental cryo-EM structures with APH-1A, however with subtle differences: The substrate-bound APH-1B γ-secretase was quite stable, but some TM helices of PS1 and APH-1B rearranged in the membrane consistent with the disorder seen in the cryo-EM data. This produces different accessibility of water molecules for the catalytic aspartates of PS1, critical for Aβ production. In particular, we find that the typical distance between the catalytic aspartates of PS1 and the C83 cleavage sites are shorter in APH-1B, that is, it represents a more closed state, due to interactions with the C-terminal fragment of PS1. Our structural-dynamic model of APH-1B alone and in γ-secretase suggests generally similar topology but some notable differences in water accessibility which may be relevant to the protein's existence in two forms and their specific function and location.
Collapse
Affiliation(s)
- Budheswar Dehury
- DTU Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Kasper P Kepp
- DTU Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
17
|
Dehury B, Tang N, Mehra R, Blundell TL, Kepp KP. Side-by-side comparison of Notch- and C83 binding to γ-secretase in a complete membrane model at physiological temperature. RSC Adv 2020; 10:31215-31232. [PMID: 35520661 PMCID: PMC9056423 DOI: 10.1039/d0ra04683c] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/15/2020] [Indexed: 12/29/2022] Open
Abstract
γ-Secretase cleaves the C99 fragment of the amyloid precursor protein, leading to formation of aggregated β-amyloid peptide central to Alzheimer's disease, and Notch, essential for cell regulation. Recent cryogenic electron microscopy (cryo-EM) structures indicate major changes upon substrate binding, a β-sheet recognition motif, and a possible helix unwinding to expose peptide bonds towards nucleophilic attack. Here we report side-by-side comparison of the 303 K dynamics of the two proteins in realistic membranes using molecular dynamics simulations. Our ensembles agree with the cryo-EM data (full-protein Cα-RMSD = 1.62–2.19 Å) but reveal distinct presenilin helix conformation states and thermal β-strand to coil transitions of C83 and Notch100. We identify distinct 303 K hydrogen bond dynamics and water accessibility of the catalytic sites. The RKRR motif (1758–1761) contributes significantly to Notch binding and serves as a “membrane anchor” that prevents Notch displacement. Water that transiently hydrogen bonds to G1753 and V1754 probably represents the catalytic nucleophile. At 303 K, Notch and C83 binding induce different conformation states, with Notch mostly present in a closed state with shorter Asp–Asp distance. This may explain the different outcome of Notch and C99 cleavage, as the latter is more imprecise with many products. Our identified conformation states may aid efforts to develop conformation-selective drugs that target C99 and Notch cleavage differently, e.g. Notch-sparing γ-secretase modulators. Distinct membrane dynamics and conformations of C83- and Notch-bound γ-secretase may aid the development of Notch-sparing treatments of Alzheimer's disease.![]()
Collapse
Affiliation(s)
- Budheswar Dehury
- Department of Chemistry, Technical University of Denmark DK-2800 Kongens Lyngby Denmark +45 45252409.,Department of Biochemistry, University of Cambridge Tennis Court Road CB2 1GA UK
| | - Ning Tang
- Department of Chemistry, Technical University of Denmark DK-2800 Kongens Lyngby Denmark +45 45252409
| | - Rukmankesh Mehra
- Department of Chemistry, Technical University of Denmark DK-2800 Kongens Lyngby Denmark +45 45252409
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge Tennis Court Road CB2 1GA UK
| | - Kasper P Kepp
- Department of Chemistry, Technical University of Denmark DK-2800 Kongens Lyngby Denmark +45 45252409
| |
Collapse
|
18
|
Mehra R, Kepp KP. Identification of Structural Calcium Binding Sites in Membrane-Bound Presenilin 1 and 2. J Phys Chem B 2020; 124:4697-4711. [PMID: 32420742 DOI: 10.1021/acs.jpcb.0c01712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Variants of presenilin (PS1 and PS2) are the main genetic risk factors of familial Alzheimer's disease and thus central to the disease etiology. Although mostly studied as catalytic units of γ-secretase controlling Aβ production, presenilins also affect calcium levels, which are disturbed in Alzheimer's disease. We investigated the interaction of calcium with both PS1 and PS2 using all-atom molecular dynamics (MD) simulations in realistic membrane models, with the specific aim to identify any Ca2+ sites. We did not observe any complete Ca2+ leak event, but we identified four persistent Ca2+ sites in membrane-bound PS1 and PS2: One in HL2 near the C-terminal of TM6, one in HL2 toward the N-terminal of TM7, a site at the catalytic aspartate on TM7, and a site at the PALP motif on TM9. The sites feature negatively charged glutamates and aspartates typical of calcium binding. Structural homology to diaspartate calcium transport proteins and mutation studies of calcium efflux support our identified calcium sites. Calcium consistently dampens HL2 motions in all comparisons (PS1, protonated PS1, PS2, protonated PS2). Due to their location in HL2 and the active site, we propose that the calcium sites control autoproteolytic maturation of presenilin by a pH-dependent conformational restriction of the HL2 recognition loop, which also regulates calcium transport proteins such as inositol 1,4,5-triphosphate receptor and ryanodine receptor. Our structural dynamics could provide a possible molecular basis for the need of both calcium and presenilin for lysosome proteolytic function, perhaps relevant also to other protein misfolding diseases.
Collapse
Affiliation(s)
- Rukmankesh Mehra
- Technical University of Denmark, DTU Chemistry, DK-2800 Kongens Lyngby, Denmark
| | - Kasper P Kepp
- Technical University of Denmark, DTU Chemistry, DK-2800 Kongens Lyngby, Denmark
| |
Collapse
|
19
|
Polychronidou E, Avramouli A, Vlamos P. Alzheimer's Disease: The Role of Mutations in Protein Folding. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1195:227-236. [PMID: 32468481 DOI: 10.1007/978-3-030-32633-3_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Misfolded proteins result when a protein follows the wrong folding pathway. Accumulation of misfolded proteins can cause disorders, known as amyloid diseases. Unfortunately, some of them are very common. The most prevalent one is Alzheimer's disease. Alzheimer's disease is a neurodegenerative disorder and the commonest form of dementia. The current study aims to assess the impact of somatic mutations in PSEN1 gene. The said mutations are the most common cause of familial Alzheimer's disease. As protein functionality can be affected by mutations, the study of possible alterations in the tertiary structure of proteins may reveal new insights related to the relationship between mutations and protein functions. To examine the effect of mutations, the primary structures and their related mutations were retrieved from public databases. Each structure (mutated and unmutated) was predicted based on effective structure prediction methodologies. A benchmarking of the structural predictive tools was accomplished. Comparative analyses of mutated and unmutated proteins were performed based on classic bioinformatics methods (TM-Score, RMSD, etc.) as well as on established shape-based descriptors retrieved from object recognition methodologies. Unsupervised methodologies were applied to the structures, in order to identify groups of mutation with similar mutational impact. Our results provide an essential knowledge toward protein's functionality in structure-based drug design.
Collapse
|
20
|
Chávez-Gutiérrez L, Szaruga M. Mechanisms of neurodegeneration - Insights from familial Alzheimer's disease. Semin Cell Dev Biol 2020; 105:75-85. [PMID: 32418657 DOI: 10.1016/j.semcdb.2020.03.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 12/18/2022]
Abstract
The rising prevalence of Alzheimer's disease (AD), together with the lack of effective treatments, portray it as one of the major health challenges of our times. Untangling AD implies advancing the knowledge of the biology that gets disrupted during the disease while deciphering the molecular and cellular mechanisms leading to AD-related neurodegeneration. In fact, a solid mechanistic understanding of the disease processes stands as an essential prerequisite for the development of safe and effective treatments. Genetics has provided invaluable clues to the genesis of the disease by revealing deterministic genes - Presenilins (PSENs) and the Amyloid Precursor Protein (APP) - that, when affected, lead in an autosomal dominant manner to early-onset, familial AD (FAD). PSEN is the catalytic subunit of the membrane-embedded γ-secretase complexes, which act as proteolytic switches regulating key cell signalling cascades. Importantly, these intramembrane proteases are responsible for the production of Amyloid β (Aβ) peptides from APP. The convergence of pathogenic mutations on one functional pathway, the amyloidogenic cleavage of APP, strongly supports the significance of this process in AD pathogenesis. Here, we review and discuss the state-of-the-art knowledge of the molecular mechanisms underlying FAD, their implications for the sporadic form of the disease and for the development of safe AD therapeutics.
Collapse
Affiliation(s)
- Lucía Chávez-Gutiérrez
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, Leuven Research Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium.
| | - Maria Szaruga
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, Leuven Research Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Kong L, Dawkins E, Campbell F, Winkler E, Derks RJE, Giera M, Kamp F, Steiner H, Kros A. Photo-controlled delivery of very long chain fatty acids to cell membranes and modulation of membrane protein function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183200. [PMID: 31972163 DOI: 10.1016/j.bbamem.2020.183200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 10/25/2022]
Abstract
The biophysical properties and biological functions of membranes are highly dependent on lipid composition. Supplementing cellular membranes with very long chain fatty acids (vlcFAs) is notoriously difficult given the extreme insolubility of vlcFAs in aqueous solution. Herein, we report a solvent-free, photochemical approach to enrich target membranes with vlcFA. To prevent aggregation of vlcFA, we created light-sensitive micelles composed exclusively of poly-ethylene-glycol-nervonic acid amphiphiles (NA-PEG), which spontaneously disassemble in the presence of lipid bilayers. Once embedded within a membrane, UV light is used to cleave off PEG, leaving free nervonic acid (NA, i.e. FA24:1) in the target membrane. When applied to living cells, free NA was processed by the cell to generate various species of membrane and other lipids with incorporated vlcFAs. In this way, we were able to alter the membrane lipid composition of cellular membranes and modulate the enzymatic activity of γ-secretase, an intramembrane protease whose dysfunction has been implicated in the onset and progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Li Kong
- Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, the Netherlands
| | - Edgar Dawkins
- Biomedical Center (BMC), Metabolic Biochemistry, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Frederick Campbell
- Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, the Netherlands
| | - Edith Winkler
- Biomedical Center (BMC), Metabolic Biochemistry, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Rico J E Derks
- Center for Proteomics & Metabolomics, Leiden University Medical Center (LUMC), the Netherlands
| | - Martin Giera
- Center for Proteomics & Metabolomics, Leiden University Medical Center (LUMC), the Netherlands
| | - Frits Kamp
- Biomedical Center (BMC), Metabolic Biochemistry, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Harald Steiner
- Biomedical Center (BMC), Metabolic Biochemistry, Ludwig-Maximilians-University, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Alexander Kros
- Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, the Netherlands.
| |
Collapse
|
22
|
Wolfe MS. Unraveling the complexity of γ-secretase. Semin Cell Dev Biol 2020; 105:3-11. [PMID: 31980377 PMCID: PMC7371508 DOI: 10.1016/j.semcdb.2020.01.005] [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: 10/07/2019] [Revised: 11/26/2019] [Accepted: 01/15/2020] [Indexed: 01/07/2023]
Abstract
γ-Secretase was initially defined as a proteolytic activity that cleaves within the transmembrane of the amyloid precursor protein (APP) to produce the amyloid β-peptide of Alzheimer's disease. The discovery of mutations in APP and the presenilins associated with familial Alzheimer's disease and their effects on APP processing dovetailed with pharmacological studies on γ-secretase, leading to the revelation that presenilins are unprecedented membrane-embedded aspartyl proteases. Other members of what became known as the γ-secretase complex were subsequently identified. In parallel with these advances, connections between presenilins and Notch receptors essential to metazoan development became evident, resulting in the concurrent realization that γ-secretase also carries out intramembrane proteolysis of Notch as part of its signaling mechanism. Substantial progress has been made toward elucidating how γ-secretase carries out complex processing of transmembrane domains, how it goes awry in familial Alzheimer's disease, the scope of its substrates, and the atomic details of its structure. Critical questions remain for future study, toward further unraveling the complexity of this unique membrane-embedded proteolytic machine and its roles in biology and disease.
Collapse
Affiliation(s)
- Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
23
|
Trambauer J, Rodríguez Sarmiento RM, Fukumori A, Feederle R, Baumann K, Steiner H. Aβ43-producing PS1 FAD mutants cause altered substrate interactions and respond to γ-secretase modulation. EMBO Rep 2020; 21:e47996. [PMID: 31762188 PMCID: PMC6945062 DOI: 10.15252/embr.201947996] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/22/2022] Open
Abstract
Abnormal generation of neurotoxic amyloid-β peptide (Aβ) 42/43 species due to mutations in the catalytic presenilin 1 (PS1) subunit of γ-secretase is the major cause of familial Alzheimer's disease (FAD). Deeper mechanistic insight on the generation of Aβ43 is still lacking, and it is unclear whether γ-secretase modulators (GSMs) can reduce the levels of this Aβ species. By comparing several types of Aβ43-generating FAD mutants, we observe that very high levels of Aβ43 are often produced when presenilin function is severely impaired. Altered interactions of C99, the precursor of Aβ, are found for all mutants and are independent of their particular effect on Aβ production. Furthermore, unlike previously described GSMs, the novel compound RO7019009 can effectively lower Aβ43 production of all mutants. Finally, substrate-binding competition experiments suggest that RO7019009 acts mechanistically after initial C99 binding. We conclude that altered C99 interactions are a common feature of diverse types of PS1 FAD mutants and that also patients with Aβ43-generating FAD mutations could in principle be treated by GSMs.
Collapse
Affiliation(s)
- Johannes Trambauer
- Biomedical Center (BMC), Metabolic BiochemistryLudwig‐Maximilians‐UniversityMunichGermany
| | | | - Akio Fukumori
- Department of Aging NeurobiologyNational Center for Geriatrics and GerontologyObuJapan
- Department of Mental Health PromotionOsaka University Graduate School of MedicineToyonakaJapan
| | - Regina Feederle
- Institute for Diabetes and Obesity, Monoclonal Antibody Core Facility, Helmholtz Center MunichGerman Research Center for Environmental HealthNeuherbergGermany
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| | - Karlheinz Baumann
- Roche Pharma Research and Early DevelopmentRoche Innovation Center Basel, F. Hoffmann‐La Roche Ltd.BaselSwitzerland
| | - Harald Steiner
- Biomedical Center (BMC), Metabolic BiochemistryLudwig‐Maximilians‐UniversityMunichGermany
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| |
Collapse
|
24
|
Mehra R, Dehury B, Kepp KP. Cryo-temperature effects on membrane protein structure and dynamics. Phys Chem Chem Phys 2020; 22:5427-5438. [DOI: 10.1039/c9cp06723j] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cryo-electron structures revolutionize biology, yet cooling effects are unclear. Using a simulation protocol of hot, cold, and rapidly cooled γ-secretase we identify cryo-contraction and modes relevant to Aβ production and cryo-analysis in general.
Collapse
Affiliation(s)
- Rukmankesh Mehra
- DTU Chemistry
- Technical University of Denmark
- DK-2800 Kongens Lyngby
- Denmark
| | - Budheswar Dehury
- DTU Chemistry
- Technical University of Denmark
- DK-2800 Kongens Lyngby
- Denmark
| | - Kasper P. Kepp
- DTU Chemistry
- Technical University of Denmark
- DK-2800 Kongens Lyngby
- Denmark
| |
Collapse
|
25
|
Computational analysis of Alzheimer-causing mutations in amyloid precursor protein and presenilin 1. Arch Biochem Biophys 2019; 678:108168. [DOI: 10.1016/j.abb.2019.108168] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/25/2019] [Accepted: 11/02/2019] [Indexed: 12/13/2022]
|
26
|
Kepp KP, Squitti R. Copper imbalance in Alzheimer’s disease: Convergence of the chemistry and the clinic. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
27
|
Dehury B, Tang N, Kepp KP. Insights into membrane-bound presenilin 2 from all-atom molecular dynamics simulations. J Biomol Struct Dyn 2019; 38:3196-3210. [PMID: 31405326 DOI: 10.1080/07391102.2019.1655481] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Presenilins 1 and 2 (PS1 or PS2) are main genetic risk factors of familial Alzheimer's disease (AD) that produce the β-amyloid (Aβ) peptides and also have important stand-alone functions related to, e.g. calcium signaling. Most work so far has focused on PS1, but humans carry both PS1 and PS2, and mutations in both cause AD. Here, we develop a computational model of PS2 in the membrane to address the question how pathogenic PS2 mutations affect the membrane-embedded protein. The models are based on cryo-electron microscopy structures of PS1 translated to PS2, augmented with missing residues and a complete all-atom membrane-water system, and equilibrated using three independent 500-ns simulations of molecular dynamics with a structure-balanced force field. We show that the nine-transmembrane channel structure is substantially controlled by major dynamics in the hydrophilic loop bridging TM6 and TM7, which functions as a 'plug' in the PS2 membrane channel. TM2, TM6, TM7 and TM9 flexibility controls the size of this channel. We find that most pathogenic PS2 mutations significantly reduce stability relative to random mutations, using a statistical ANOVA test with all possible mutations in the affected sites as a control. The associated loss of compactness may also impair calcium affinity. Remarkably, similar properties of the open state are known to impair the binding of substrates to γ-secretase, and we thus argue that the two mechanisms could be functionally related.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Budheswar Dehury
- DTU Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ning Tang
- DTU Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Kasper P Kepp
- DTU Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
28
|
Dehury B, Tang N, Blundell TL, Kepp KP. Structure and dynamics of γ-secretase with presenilin 2 compared to presenilin 1. RSC Adv 2019; 9:20901-20916. [PMID: 35515530 PMCID: PMC9065803 DOI: 10.1039/c9ra02623a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/27/2019] [Indexed: 12/12/2022] Open
Abstract
Severe early-onset familial Alzheimer's disease (FAD) is caused by more than 200 different mutations in the genes coding for presenilin, the catalytic subunit of the 4-subunit protease complex γ-secretase, which cleaves the C99 fragment of the amyloid precursor protein (APP) to produce Aβ peptides. γ-Secretase exists with either of two homologues, PS1 and PS2. All cryo-electron microscopic structures and computational work has so far focused on γ-secretase with PS1, yet PS2 mutations also cause FAD. A central question is thus whether there are structural and dynamic differences between PS1 and PS2. To address this question, we use the cryo-electron microscopic data for PS1 to develop the first structural and dynamic model of PS2-γ-secretase in the catalytically relevant mature membrane-bound state at ambient temperature, equilibrated by three independent 500 ns molecular dynamics simulations. We find that the characteristic nicastrin extra-cellular domain breathing mode and major movements in the cytosolic loop between TM6 and TM7 occur in both PS2- and PS1-γ-secretase. The overall structures and conformational states are similar, suggesting similar catalytic activities. However, at the sequence level, charge-controlled membrane-anchoring is extracellular for PS1 and intracellular for PS2, which suggests different subcellular locations. The tilt angles of the TM2, TM6, TM7 and TM9 helices differ in the two forms of γ-secretase, suggesting that the two proteins have somewhat different substrate processing and channel sizes. Our MD simulations consistently indicated that PS2 retains several water molecules near the catalytic site at the bilayer, as required for catalysis. The possible reasons for the differences of PS1 and PS2 are discussed in relation to their location and function.
Collapse
Affiliation(s)
- Budheswar Dehury
- Department of Chemistry, Technical University of Denmark DK-2800 Kongens Lyngby Denmark +045 45252409
| | - Ning Tang
- Department of Chemistry, Technical University of Denmark DK-2800 Kongens Lyngby Denmark +045 45252409
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge Cambridge CB2 1GA UK
| | - Kasper P Kepp
- Department of Chemistry, Technical University of Denmark DK-2800 Kongens Lyngby Denmark +045 45252409
| |
Collapse
|
29
|
Blanco JA, Alonso A, Blanco J, Rojo E, Tellería JJ, Torres MA, Uribe F. Novel presenilin 1 mutation (p.Thr-Pro116-117Ser-Thr) in a Spanish family with early-onset Alzheimer's disease. Neurobiol Aging 2019; 84:238.e19-238.e24. [PMID: 31204041 DOI: 10.1016/j.neurobiolaging.2019.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 11/26/2022]
Abstract
Presenilin 1 (PSEN1) is a γ-secretase component, which is in charge of the amyloid precursor protein (APP) cleavage. APP is believed to play a central role in the pathogenesis of Alzheimer's disease (AD). PSEN1 mutations are the most important causes of familial AD, being related to the earlier onset and rapid progression of the disease. Presenilins and APP mutations represent an extraordinary opportunity to study the pathophysiology of AD. We describe the clinical and genetic study of a 37-year-old male patient with a novel mutation in PSEN1 (p.Thr-Pro116-117Ser-Thr). We have studied the pedigree of his family with a further 9 members affected, all of them with onset in their 30s. We have also described the clinical data and results of brain biopsies in 2 of them. DNA sequencing of a tissue sample from an uncle of the patient, who died of AD in the 80s, showed the same mutation as in the patient. These data and predictive analysis indicate the pathogenicity of the mutation.
Collapse
Affiliation(s)
- Jose Antonio Blanco
- Psychiatry Service, Clinical University Hospital of Valladolid, Valladolid, Spain; School of Medicine, University of Valladolid, Valladolid, Spain.
| | - Adrian Alonso
- Psychiatry Service, Clinical University Hospital of Valladolid, Valladolid, Spain
| | - Jorge Blanco
- Psychiatry Service, Basurto Hospital of Bilbao, Bilbao, Spain
| | - Esther Rojo
- Neurology Service, Clinical University Hospital of Valladolid, Valladolid, Spain
| | | | - Maria Angeles Torres
- Histopathology Service, Rio Hortega University Hospital of Valladolid, Valladolid, Spain
| | - Fernando Uribe
- Psychiatry Service, Clinical University Hospital of Valladolid, Valladolid, Spain
| |
Collapse
|
30
|
Molecular dynamics of C99-bound γ-secretase reveal two binding modes with distinct compactness, stability, and active-site retention: implications for Aβ production. Biochem J 2019; 476:1173-1189. [PMID: 30910800 DOI: 10.1042/bcj20190023] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022]
Abstract
The membrane protease γ-secretase cleaves the C99 fragment of the amyloid precursor protein, thus producing the Aβ peptides central to Alzheimer's disease. Cryo-electron microscopy has provided the topology but misses the membrane and loop parts that contribute to substrate binding. We report here an essentially complete atomic model of C99 within wild-type γ-secretase that respects all the experimental constraints and additionally describes loop, helix, and C99 substrate dynamics in a realistic all-atom membrane. Our model represents the matured auto-cleaved state required for catalysis. From two independent 500-ns molecular dynamic simulations, we identify two conformation states of C99 in equilibrium, a compact and a loose state. Our simulations provide a basis for C99 processing and Aβ formation and explain the production of longer and shorter Aβ, as the compact state retains C99 for longer and thus probably trims to shorter Aβ peptides. We expect pathogenic presenilin mutations to stabilize the loose over the compact state. The simulations detail the role of the Lys53-Lys54-Lys55 anchor for C99 binding, a loss of helicity of bound C99, and positioning of Thr48 and Leu49 leading to alternative trimming pathways on opposite sides of the C99 helix in three amino acid steps. The C99 binding topology resembles that of C83-bound γ-secretase without membrane but lacks a presenilin 1-C99 β-sheet, which could be induced by C83's stronger binding. The loose state should be selectively disfavored by γ-secretase modulators to increase C99 trimming and reduce the formation of longer Aβ, a strategy that is currently much explored but has lacked a structural basis.
Collapse
|
31
|
Tang N, Dehury B, Kepp KP. Computing the Pathogenicity of Alzheimer’s Disease Presenilin 1 Mutations. J Chem Inf Model 2019; 59:858-870. [DOI: 10.1021/acs.jcim.8b00896] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ning Tang
- Department of Chemistry, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Budheswar Dehury
- Department of Chemistry, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Kasper P. Kepp
- Department of Chemistry, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| |
Collapse
|