1
|
Schrempel S, Kottwitz AK, Piechotta A, Gnoth K, Büschgens L, Hartlage-Rübsamen M, Morawski M, Schenk M, Kleinschmidt M, Serrano GE, Beach TG, Rostagno A, Ghiso J, Heneka MT, Walter J, Wirths O, Schilling S, Roßner S. Identification of isoAsp7-Aβ as a major Aβ variant in Alzheimer's disease, dementia with Lewy bodies and vascular dementia. Acta Neuropathol 2024; 148:78. [PMID: 39625512 PMCID: PMC11615120 DOI: 10.1007/s00401-024-02824-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 12/06/2024]
Abstract
The formation of amyloid-β (Aβ) aggregates in brain is a neuropathological hallmark of Alzheimer's disease (AD). However, there is mounting evidence that Aβ also plays a pathogenic role in other types of dementia and that specific post-translational Aβ modifications contribute to its pathogenic profile. The objective of this study was to test the hypothesis that distinct types of dementia are characterized by specific patterns of post-translationally modified Aβ variants. We conducted a comparative analysis and quantified Aβ as well as Aβ with pyroglutamate (pGlu3-Aβ and pGlu11-Aβ), N-truncation (Aβ(4-X)), isoaspartate racemization (isoAsp7-Aβ and isoAsp27-Aβ), phosphorylation (pSer8-Aβ and pSer26-Aβ) or nitration (3NTyr10-Aβ) modification in post mortem human brain tissue from non-demented control subjects in comparison to tissue classified as pre-symptomatic AD (Pre-AD), AD, dementia with Lewy bodies and vascular dementia. Aβ modification-specific immunohistochemical labelings of brain sections from the posterior superior temporal gyrus were examined by machine learning-based segmentation protocols and immunoassay analyses in brain tissue after sequential Aβ extraction were carried out. Our findings revealed that AD cases displayed the highest concentrations of all Aβ variants followed by dementia with Lewy bodies, Pre-AD, vascular dementia and non-demented controls. With both analytical methods, we identified the isoAsp7-Aβ variant as a highly abundant Aβ form in all clinical conditions, followed by Aβ(4-X), pGlu3-Aβ, pGlu11-Aβ and pSer8-Aβ. These Aβ variants were detected in distinct plaque types of compact, coarse-grained, cored and diffuse morphologies and, with varying frequencies, in cerebral blood vessels. The 3NTyr10-Aβ, pSer26-Aβ and isoAsp27-Aβ variants were not found to be present in Aβ plaques but were detected intraneuronally. There was a strong positive correlation between isoAsp7-Aβ and Thal phase and a moderate negative correlation between isoAsp7-Aβ and performance on the Mini Mental State Examination. Furthermore, the abundance of all Aβ variants was highest in APOE 3/4 carriers. In aggregation assays, the isoAsp7-Aβ, pGlu3-Aβ and pGlu11-Aβ variants showed instant fibril formation without lag phase, whereas Aβ(4-X), pSer26-Aβ and isoAsp27-Aβ did not form fibrils. We conclude that targeting Aβ post-translational modifications, and in particular the highly abundant isoAsp7-Aβ variant, might be considered for diagnostic and therapeutic approaches in different types of dementia. Hence, our findings might have implications for current antibody-based therapies of AD.
Collapse
Affiliation(s)
- Sarah Schrempel
- Paul Flechsig Institute - Centre of Neuropathology and Brain Research, University of Leipzig, Liebigstraße 19, 04103, Leipzig, Germany
| | - Anna Katharina Kottwitz
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120, Halle (Saale), Germany
- Center for Natural Product-based Therapeutics, Anhalt University of Applied Sciences, 06366, Köthen, Germany
| | - Anke Piechotta
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120, Halle (Saale), Germany
| | - Kathrin Gnoth
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120, Halle (Saale), Germany
- Center for Natural Product-based Therapeutics, Anhalt University of Applied Sciences, 06366, Köthen, Germany
| | - Luca Büschgens
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, 37075, Göttingen, Germany
| | - Maike Hartlage-Rübsamen
- Paul Flechsig Institute - Centre of Neuropathology and Brain Research, University of Leipzig, Liebigstraße 19, 04103, Leipzig, Germany
| | - Markus Morawski
- Paul Flechsig Institute - Centre of Neuropathology and Brain Research, University of Leipzig, Liebigstraße 19, 04103, Leipzig, Germany
| | - Mathias Schenk
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120, Halle (Saale), Germany
| | - Martin Kleinschmidt
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120, Halle (Saale), Germany
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Brain and Body Donation Program, Banner Sun Health Research Institute, 10515 W Santa Fe Drive, Sun City, AZ, 85351, USA
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Brain and Body Donation Program, Banner Sun Health Research Institute, 10515 W Santa Fe Drive, Sun City, AZ, 85351, USA
| | - Agueda Rostagno
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Jorge Ghiso
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | - Jochen Walter
- Center of Neurology, Molecular Cell Biology, University Hospital Bonn, 53127, Bonn, Germany
| | - Oliver Wirths
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, 37075, Göttingen, Germany
| | - Stephan Schilling
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120, Halle (Saale), Germany
- Center for Natural Product-based Therapeutics, Anhalt University of Applied Sciences, 06366, Köthen, Germany
| | - Steffen Roßner
- Paul Flechsig Institute - Centre of Neuropathology and Brain Research, University of Leipzig, Liebigstraße 19, 04103, Leipzig, Germany.
| |
Collapse
|
2
|
Kenyaga JM, Qiang W. Extraction of In-Cell β-Amyloid Fibrillar Aggregates for Studying Molecular-Level Structural Propagations Using Solid-State NMR Spectroscopy. Biochemistry 2024; 63:2557-2564. [PMID: 39348718 DOI: 10.1021/acs.biochem.4c00395] [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] [Indexed: 10/02/2024]
Abstract
Molecular-level structural polymorphisms of β-amyloid (Aβ) fibrils have recently been recognized as pathologically significant. High-resolution solid-state nuclear magnetic resonance (ssNMR) spectroscopy has been utilized to study these structural polymorphisms, particularly in ex-vivo fibrils seeded from amyloid extracts of post-mortem brain tissues of Alzheimer's disease (AD) patients. One unaddressed question in current ex-vivo seeding protocol is whether fibrillation from exogenous monomeric Aβ peptides, added to the extracted seeds, can be quantitatively suppressed. Addressing this issue is critical because uncontrolled fibrillation could introduce biased molecular structural polymorphisms in the resulting fibrils. Here, we present a workflow to optimize the key parameters of ex-vivo seeding protocols, focusing on the quantification of amyloid extraction and the selection of exogenous monomeric Aβ concentrations to minimize nonseeded fibrillation. We validate this workflow using three structurally different 40-residue Aβ (Aβ40) fibrillar seeds, demonstrating their ability to propagate their structural features to exogenous wild-type Aβ40.
Collapse
Affiliation(s)
- June M Kenyaga
- Department of Chemistry, Binghamton University, the State University of New York, Vestal, New York 13850, United States
| | - Wei Qiang
- Department of Chemistry, Binghamton University, the State University of New York, Vestal, New York 13850, United States
| |
Collapse
|
3
|
Rodríguez-Flórez LV, González-Marcos M, García-Mingüens E, Retamosa MDG, Kawase M, Selva E, Sansano JM. Phosphine Catalyzed Michael-Type Additions: The Synthesis of Glutamic Acid Derivatives from Arylidene- α-amino Esters. Molecules 2024; 29:342. [PMID: 38257255 PMCID: PMC10820836 DOI: 10.3390/molecules29020342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 12/28/2023] [Accepted: 01/07/2024] [Indexed: 01/24/2024] Open
Abstract
The reaction of arylidene-α-amino esters with electrophilic alkenes to yield Michael-type addition compounds is optimized using several phosphines as organocatalysts. The transformation is very complicated due to the generation of several final compounds, including those derived from the 1,3-dipolar cycloadditions. For this reason, the selection of the reaction conditions is a very complex task and the slow addition of the acrylic system is very important to complete the process. The study of the variation in the structural components of the starting imino ester is performed as well as the expansion of other electron-poor alkenes. The crude products have a purity higher than 90% in most cases without any purification. A plausible mechanism is detailed based on the bibliography and the experimental results. The synthesis of pyroglutamate entities, after the reduction of the imino group and cyclization, is performed in high yields. In addition, the hydrolysis of the imino group, under acidic media, represents a direct access to glutamate surrogates.
Collapse
Affiliation(s)
- Lesly V. Rodríguez-Flórez
- Departamento de Química Orgánica, Centro de Innovación en Química Avanzada (ORFEO-CINQA) and Instituto de Síntesis Orgánica, Universidad de Alicante, Ctra. Alicante-San Vicente s/n, 03080 Alicante, Spain
| | - María González-Marcos
- Departamento de Química Orgánica, Centro de Innovación en Química Avanzada (ORFEO-CINQA) and Instituto de Síntesis Orgánica, Universidad de Alicante, Ctra. Alicante-San Vicente s/n, 03080 Alicante, Spain
| | - Eduardo García-Mingüens
- Medalchemy, S. L. Ancha de Castelar, 46-48, entlo. A. San Vicente del Raspeig, 03690 Alicante, Spain
| | - María de Gracia Retamosa
- Departamento de Química Orgánica, Centro de Innovación en Química Avanzada (ORFEO-CINQA) and Instituto de Síntesis Orgánica, Universidad de Alicante, Ctra. Alicante-San Vicente s/n, 03080 Alicante, Spain
| | - Misa Kawase
- Departamento de Química Orgánica, Centro de Innovación en Química Avanzada (ORFEO-CINQA) and Instituto de Síntesis Orgánica, Universidad de Alicante, Ctra. Alicante-San Vicente s/n, 03080 Alicante, Spain
| | - Elisabet Selva
- Medalchemy, S. L. Ancha de Castelar, 46-48, entlo. A. San Vicente del Raspeig, 03690 Alicante, Spain
| | - José M. Sansano
- Departamento de Química Orgánica, Centro de Innovación en Química Avanzada (ORFEO-CINQA) and Instituto de Síntesis Orgánica, Universidad de Alicante, Ctra. Alicante-San Vicente s/n, 03080 Alicante, Spain
| |
Collapse
|
4
|
Park JH, Iwamoto M, Yun JH, Uchikubo-Kamo T, Son D, Jin Z, Yoshida H, Ohki M, Ishimoto N, Mizutani K, Oshima M, Muramatsu M, Wakita T, Shirouzu M, Liu K, Uemura T, Nomura N, Iwata S, Watashi K, Tame JRH, Nishizawa T, Lee W, Park SY. Structural insights into the HBV receptor and bile acid transporter NTCP. Nature 2022; 606:1027-1031. [PMID: 35580630 PMCID: PMC9242859 DOI: 10.1038/s41586-022-04857-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 05/11/2022] [Indexed: 01/05/2023]
Abstract
Around 250 million people are infected with hepatitis B virus (HBV) worldwide1, and 15 million may also carry the satellite virus hepatitis D virus (HDV), which confers even greater risk of severe liver disease2. The HBV receptor has been identified as sodium taurocholate co-transporting polypeptide (NTCP), which interacts directly with the first 48 amino acid residues of the N-myristoylated N-terminal preS1 domain of the viral large protein3. Despite the pressing need for therapeutic agents to counter HBV, the structure of NTCP remains unsolved. This 349-residue protein is closely related to human apical sodium-dependent bile acid transporter (ASBT), another member of the solute carrier family SLC10. Crystal structures have been reported of similar bile acid transporters from bacteria4,5, and these models are believed to resemble closely both NTCP and ASBT. Here we have used cryo-electron microscopy to solve the structure of NTCP bound to an antibody, clearly showing that the transporter has no equivalent of the first transmembrane helix found in other SLC10 proteins, and that the N terminus is exposed on the extracellular face. Comparison of our structure with those of related proteins indicates a common mechanism of bile acid transport, but the NTCP structure displays an additional pocket formed by residues that are known to interact with preS1, presenting new opportunities for structure-based drug design. Cryo-electron structures of the hepatitis B virus receptor NTCP show a distinct membrane topology compared with other SLC10 proteins, but a common bile acid transport mechanism that is shared with related mammalian and bacterial proteins.
Collapse
Affiliation(s)
- Jae-Hyun Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Masashi Iwamoto
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ji-Hye Yun
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.,PCG-Biotech, Seoul, South Korea
| | - Tomomi Uchikubo-Kamo
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Donghwan Son
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Zeyu Jin
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.,Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Hisashi Yoshida
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Mio Ohki
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Naito Ishimoto
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Kenji Mizutani
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Mizuki Oshima
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Biological Sciences, Tokyo University of Science, Noda, Japan
| | - Masamichi Muramatsu
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Kehong Liu
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Uemura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norimichi Nomura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - So Iwata
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,RIKEN SPring-8 Center, Sayo-gun, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Biological Sciences, Tokyo University of Science, Noda, Japan.,Research Center for Drug and Vaccine Development, Tokyo, Japan
| | - Jeremy R H Tame
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Tomohiro Nishizawa
- Laboratory of Biomembrane Dynamics, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Weontae Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea. .,PCG-Biotech, Seoul, South Korea.
| | - Sam-Yong Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.
| |
Collapse
|
5
|
Gutiérrez-Fernández J, Javaid F, De Rossi G, Chudasama V, Greenwood J, Moss SE, Luecke H. Structural basis of human LRG1 recognition by Magacizumab, a humanized monoclonal antibody with therapeutic potential. Acta Crystallogr D Struct Biol 2022; 78:725-734. [PMID: 35647920 PMCID: PMC9159282 DOI: 10.1107/s2059798322004132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 04/19/2022] [Indexed: 11/10/2022] Open
Abstract
The formation of new dysfunctional blood vessels is a crucial stage in the development of various conditions such as macular degeneration, diabetes, cardiovascular disease, neurological disease and inflammatory disorders, as well as during tumor growth, eventually contributing to metastasis. An important factor involved in pathogenic angiogenesis is leucine-rich α-2-glycoprotein 1 (LRG1), the antibody blockade of which has been shown to lead to a reduction in both choroidal neovascularization and tumor growth in mouse models. In this work, the structural interactions between the LRG1 epitope and the Fab fragment of Magacizumab, a humanized function-blocking IgG4 against LRG1, are analysed, determining its specific binding mode and the key residues involved in LRG1 recognition. Based on these structural findings, a series of mutations are suggested that could be introduced into Magacizumab to increase its affinity for LRG1, as well as a model of the entire Fab-LRG1 complex that could enlighten new strategies to enhance affinity, consequently leading towards an even more efficient therapeutic.
Collapse
Affiliation(s)
- Javier Gutiérrez-Fernández
- Structural Biology and Drug Discovery Group, Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway
| | - Faiza Javaid
- Institute of Ophthalmology, University College London, London, United Kingdom
- Department of Chemistry, University College London, London, United Kingdom
| | - Giulia De Rossi
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Vijay Chudasama
- Department of Chemistry, University College London, London, United Kingdom
| | - John Greenwood
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Stephen E. Moss
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Hartmut Luecke
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
6
|
Marengo L, Armbrust F, Schoenherr C, Storck SE, Schmitt U, Zampar S, Wirths O, Altmeppen H, Glatzel M, Kaether C, Weggen S, Becker-Pauly C, Pietrzik CU. Meprin β knockout reduces brain Aβ levels and rescues learning and memory impairments in the APP/lon mouse model for Alzheimer's disease. Cell Mol Life Sci 2022; 79:168. [PMID: 35235058 PMCID: PMC8891209 DOI: 10.1007/s00018-022-04205-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/26/2022] [Accepted: 02/10/2022] [Indexed: 11/25/2022]
Abstract
β-Site amyloid precursor protein (APP) cleaving enzyme-1 (BACE1) is the major described β-secretase to generate Aβ peptides in Alzheimer's disease (AD). However, all therapeutic attempts to block BACE1 activity and to improve AD symptoms have so far failed. A potential candidate for alternative Aβ peptides generation is the metalloproteinase meprin β, which cleaves APP predominantly at alanine in p2 and in this study we can detect an increased meprin β expression in AD brain. Here, we report the generation of the transgenic APP/lon mouse model of AD lacking the functional Mep1b gene (APP/lon × Mep1b-/-). We examined levels of canonical and truncated Aβ species using urea-SDS-PAGE, ELISA and immunohistochemistry in brains of APP/lon mouse × Mep1b-/-. Additionally, we investigated the cognitive abilities of these mice during the Morris water maze task. Aβ1-40 and 1-42 levels are reduced in APP/lon mice when meprin β is absent. Immunohistochemical staining of mouse brain sections revealed that N-terminally truncated Aβ2-x peptide deposition is decreased in APP/lon × Mep1b-/- mice. Importantly, loss of meprin β improved cognitive abilities and rescued learning behavior impairments in APP/lon mice. These observations indicate an important role of meprin β within the amyloidogenic pathway and Aβ production in vivo.
Collapse
Affiliation(s)
- Liana Marengo
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Fred Armbrust
- Institute of Biochemistry, Unit for Degradomics of the Protease Web, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Caroline Schoenherr
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Steffen E. Storck
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ulrich Schmitt
- Leibniz-Institute for Resilience Research, Mainz, Germany
| | - Silvia Zampar
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Oliver Wirths
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Hermann Altmeppen
- Institute of Neuropathology, University Medical Center HH-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center HH-Eppendorf, Hamburg, Germany
| | | | - Sascha Weggen
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Christoph Becker-Pauly
- Institute of Biochemistry, Unit for Degradomics of the Protease Web, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Claus U. Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Duesbergweg 6, 55099 Mainz, Germany
| |
Collapse
|
7
|
Discovery of a novel pseudo β-hairpin structure of N-truncated amyloid-β for use as a vaccine against Alzheimer's disease. Mol Psychiatry 2022; 27:840-848. [PMID: 34776512 DOI: 10.1038/s41380-021-01385-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/16/2021] [Accepted: 10/27/2021] [Indexed: 12/21/2022]
Abstract
One of the hallmarks of Alzheimer's disease (AD) are deposits of amyloid-beta (Aβ) protein in amyloid plaques in the brain. The Aβ peptide exists in several forms, including full-length Aβ1-42 and Aβ1-40 - and the N-truncated species, pyroglutamate Aβ3-42 and Aβ4-42, which appear to play a major role in neurodegeneration. We previously identified a murine antibody (TAP01), which binds specifically to soluble, non-plaque N-truncated Aβ species. By solving crystal structures for TAP01 family antibodies bound to pyroglutamate Aβ3-14, we identified a novel pseudo β-hairpin structure in the N-terminal region of Aβ and show that this underpins its unique binding properties. We engineered a stabilised cyclic form of Aβ1-14 (N-Truncated Amyloid Peptide AntibodieS; the 'TAPAS' vaccine) and showed that this adopts the same 3-dimensional conformation as the native sequence when bound to TAP01. Active immunisation of two mouse models of AD with the TAPAS vaccine led to a striking reduction in amyloid-plaque formation, a rescue of brain glucose metabolism, a stabilisation in neuron loss, and a rescue of memory deficiencies. Treating both models with the humanised version of the TAP01 antibody had similar positive effects. Here we report the discovery of a unique conformational epitope in the N-terminal region of Aβ, which offers new routes for active and passive immunisation against AD.
Collapse
|
8
|
Pyroglutamate Aβ cascade as drug target in Alzheimer's disease. Mol Psychiatry 2022; 27:1880-1885. [PMID: 34880449 PMCID: PMC9126800 DOI: 10.1038/s41380-021-01409-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/19/2021] [Accepted: 11/25/2021] [Indexed: 02/07/2023]
Abstract
One of the central aims in Alzheimer's disease (AD) research is the identification of clinically relevant drug targets. A plethora of potential molecular targets work very well in preclinical model systems both in vitro and in vivo in AD mouse models. However, the lack of translation into clinical settings in the AD field is a challenging endeavor. Although it is long known that N-terminally truncated and pyroglutamate-modified Abeta (AβpE3) peptides are abundantly present in the brain of AD patients, form stable and soluble low-molecular weight oligomers, and induce neurodegeneration in AD mouse models, their potential as drug target has not been generally accepted in the past. This situation has dramatically changed with the report that passive immunization with donanemab, an AβpE3-specific antibody, cleared aymloid plaques and stabilized cognitive deficits in a group of patients with mild AD in a phase II trial. This review summarizes the current knowledge on the molecular mechanisms of generation of AβpE, its biochemical properties, and the intervention points as a drug target in AD.
Collapse
|
9
|
Hoffmann T, Rahfeld JU, Schenk M, Ponath F, Makioka K, Hutter-Paier B, Lues I, Lemere CA, Schilling S. Combination of the Glutaminyl Cyclase Inhibitor PQ912 (Varoglutamstat) and the Murine Monoclonal Antibody PBD-C06 (m6) Shows Additive Effects on Brain Aβ Pathology in Transgenic Mice. Int J Mol Sci 2021; 22:ijms222111791. [PMID: 34769222 PMCID: PMC8584206 DOI: 10.3390/ijms222111791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/07/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
Compelling evidence suggests that pyroglutamate-modified Aβ (pGlu3-Aβ; AβN3pG) peptides play a pivotal role in the development and progression of Alzheimer’s disease (AD). Approaches targeting pGlu3-Aβ by glutaminyl cyclase (QC) inhibition (Varoglutamstat) or monoclonal antibodies (Donanemab) are currently in clinical development. Here, we aimed at an assessment of combination therapy of Varoglutamstat (PQ912) and a pGlu3-Aβ-specific antibody (m6) in transgenic mice. Whereas the single treatments at subtherapeutic doses show moderate (16–41%) but statistically insignificant reduction of Aβ42 and pGlu-Aβ42 in mice brain, the combination of both treatments resulted in significant reductions of Aβ by 45–65%. Evaluation of these data using the Bliss independence model revealed a combination index of ≈1, which is indicative for an additive effect of the compounds. The data are interpreted in terms of different pathways, in which the two drugs act. While PQ912 prevents the formation of pGlu3-Aβ in different compartments, the antibody is able to clear existing pGlu3-Aβ deposits. The results suggest that combination of the small molecule Varoglutamstat and a pE3Aβ-directed monoclonal antibody may allow a reduction of the individual compound doses while maintaining the therapeutic effect.
Collapse
Affiliation(s)
- Torsten Hoffmann
- Vivoryon Therapeutics N.V., Weinbergweg 22, 06120 Halle, Germany;
- Correspondence: (T.H.); (S.S.)
| | - Jens-Ulrich Rahfeld
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, Weinbergweg 22, 06120 Halle, Germany; (J.-U.R.); (M.S.)
| | - Mathias Schenk
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, Weinbergweg 22, 06120 Halle, Germany; (J.-U.R.); (M.S.)
| | - Falk Ponath
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Rd., Boston, MA 02115, USA; (F.P.); (K.M.); (C.A.L.)
| | - Koki Makioka
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Rd., Boston, MA 02115, USA; (F.P.); (K.M.); (C.A.L.)
| | - Birgit Hutter-Paier
- QPS Austria GmbH, Department of Neuropharmacology, Parkring 12, A-8074 Grambach, Austria;
| | - Inge Lues
- Vivoryon Therapeutics N.V., Weinbergweg 22, 06120 Halle, Germany;
| | - Cynthia A. Lemere
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Rd., Boston, MA 02115, USA; (F.P.); (K.M.); (C.A.L.)
| | - Stephan Schilling
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, Weinbergweg 22, 06120 Halle, Germany; (J.-U.R.); (M.S.)
- Anhalt University of Applied Sciences, Bernburger Straße 55, 06366 Köthen, Germany
- Correspondence: (T.H.); (S.S.)
| |
Collapse
|
10
|
Hu ZW, Cruceta L, Zhang S, Sun Y, Qiang W. Cross-Seeded Fibrillation Induced by Pyroglutamate-3 and Truncated Aβ 40 Variants Leads to Aβ 40 Structural Polymorphism Modulation and Elevated Toxicity. ACS Chem Neurosci 2021; 12:3625-3637. [PMID: 34524791 DOI: 10.1021/acschemneuro.1c00341] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The pathological amyloid plaques in Alzheimer's disease (AD) patients contain not only the wild-type β-amyloid (wt-Aβ) peptide sequences but also a variety of post-translationally modified variants. The pyroglutamate-3 Aβ (pyroE3-Aβ), which is generated from its truncated precursors ΔE3-Aβ, shows the highest abundance among all modified Aβ variants. Previous works have shown that pyroE3-Aβ and/or ΔE3-Aβ, compared with the wild-type sequences, led to a more rapid fibrillation process and final fibrils with higher neuronal cytotoxicity levels. However, much less is known about how the formation of pyroE3/ΔE3-Aβ fibrils would affect the amyloid deposition of wt-Aβ peptides, which are the main pathological events in AD. We show in the present work that the pyroE3/ΔE3-Aβ40 fibrils differ significantly from the wt-Aβ40 fibrils in terms of their molecular structures. When added into monomeric wt-Aβ40 peptides, these variant fibrils can cross-seed the formation of wt-Aβ40 fibrils with fibrillation kinetics that are greater than the self-seeded fibrillation of wt-Aβ40. Furthermore, the cross-seeding process modulates the molecular structures of the yielded wt-Aβ40 fibrils, which show similar features as their variant seeds. The cross-seeded fibrillation process also induces higher cytotoxicity levels compared with the self-seeded fibrillation of wt-Aβ40. Overall, our results support the hypothesis that pyroE3 and ΔE3-Aβ40 variants may serve as triggering factors of the pathological amyloid aggregation of wt-Aβ40 and may underlie the pathological significance of pyroE3/ΔE3-Aβ40 variants on the structural polymorphism of Aβ deposits.
Collapse
Affiliation(s)
- Zhi-Wen Hu
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Letticia Cruceta
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Shiyue Zhang
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Yan Sun
- Small Scale Systems Integration and Package (S3IP) Center, Binghamton University, Binghamton, New York 13902, United States
| | - Wei Qiang
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| |
Collapse
|
11
|
Hartlage-Rübsamen M, Bluhm A, Moceri S, Machner L, Köppen J, Schenk M, Hilbrich I, Holzer M, Weidenfeller M, Richter F, Coras R, Serrano GE, Beach TG, Schilling S, von Hörsten S, Xiang W, Schulze A, Roßner S. A glutaminyl cyclase-catalyzed α-synuclein modification identified in human synucleinopathies. Acta Neuropathol 2021; 142:399-421. [PMID: 34309760 PMCID: PMC8357657 DOI: 10.1007/s00401-021-02349-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 12/22/2022]
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder that is neuropathologically characterized by degeneration of dopaminergic neurons of the substantia nigra (SN) and formation of Lewy bodies and Lewy neurites composed of aggregated α-synuclein. Proteolysis of α-synuclein by matrix metalloproteinases was shown to facilitate its aggregation and to affect cell viability. One of the proteolysed fragments, Gln79-α-synuclein, possesses a glutamine residue at its N-terminus. We argue that glutaminyl cyclase (QC) may catalyze the pyroglutamate (pGlu)79-α-synuclein formation and, thereby, contribute to enhanced aggregation and compromised degradation of α-synuclein in human synucleinopathies. Here, the kinetic characteristics of Gln79-α-synuclein conversion into the pGlu-form by QC are shown using enzymatic assays and mass spectrometry. Thioflavin T assays and electron microscopy demonstrated a decreased potential of pGlu79-α-synuclein to form fibrils. However, size exclusion chromatography and cell viability assays revealed an increased propensity of pGlu79-α-synuclein to form oligomeric aggregates with high neurotoxicity. In brains of wild-type mice, QC and α-synuclein were co-expressed by dopaminergic SN neurons. Using a specific antibody against the pGlu-modified neo-epitope of α-synuclein, pGlu79-α-synuclein aggregates were detected in association with QC in brains of two transgenic mouse lines with human α-synuclein overexpression. In human brain samples of PD and dementia with Lewy body subjects, pGlu79-α-synuclein was shown to be present in SN neurons, in a number of Lewy bodies and in dystrophic neurites. Importantly, there was a spatial co-occurrence of pGlu79-α-synuclein with the enzyme QC in the human SN complex and a defined association of QC with neuropathological structures. We conclude that QC catalyzes the formation of oligomer-prone pGlu79-α-synuclein in human synucleinopathies, which may—in analogy to pGlu-Aβ peptides in Alzheimer’s disease—act as a seed for pathogenic protein aggregation.
Collapse
|
12
|
John A, Järvå MA, Shah S, Mao R, Chappaz S, Birkinshaw RW, Czabotar PE, Lo AW, Scott NE, Goddard-Borger ED. Yeast- and antibody-based tools for studying tryptophan C-mannosylation. Nat Chem Biol 2021; 17:428-437. [PMID: 33542533 DOI: 10.1038/s41589-020-00727-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 12/21/2020] [Indexed: 01/30/2023]
Abstract
Tryptophan C-mannosylation is an unusual co-translational protein modification performed by metazoans and apicomplexan protists. The prevalence and biological functions of this modification are poorly understood, with progress in the field hampered by a dearth of convenient tools for installing and detecting the modification. Here, we engineer a yeast system to produce a diverse array of proteins with and without tryptophan C-mannosylation and interrogate the modification's influence on protein stability and function. This system also enabled mutagenesis studies to identify residues of the glycosyltransferase and its protein substrates that are crucial for catalysis. The collection of modified proteins accrued during this work facilitated the generation and thorough characterization of monoclonal antibodies against tryptophan C-mannosylation. These antibodies empowered proteomic analyses of the brain C-glycome by enriching for peptides possessing tryptophan C-mannosylation. This study revealed many new modification sites on proteins throughout the secretory pathway with both conventional and non-canonical consensus sequences.
Collapse
Affiliation(s)
- Alan John
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Michael A Järvå
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Sayali Shah
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Runyu Mao
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Stephane Chappaz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Richard W Birkinshaw
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Alvin W Lo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Ethan D Goddard-Borger
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
13
|
Neddens J, Daurer M, Flunkert S, Beutl K, Loeffler T, Walker L, Attems J, Hutter-Paier B. Correlation of pyroglutamate amyloid β and ptau Ser202/Thr205 levels in Alzheimer's disease and related murine models. PLoS One 2020; 15:e0235543. [PMID: 32645028 PMCID: PMC7347153 DOI: 10.1371/journal.pone.0235543] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/17/2020] [Indexed: 01/14/2023] Open
Abstract
Senile plaques frequently contain Aβ-pE(3), a N-terminally truncated Aβ species that is more closely linked to AD compared to other Aβ species. Tau protein is highly phosphorylated at several residues in AD, and specifically phosphorylation at Ser202/Thr205 is known to be increased in AD. Several studies suggest that formation of plaques and tau phosphorylation might be linked to each other. To evaluate if Aβ-pE(3) and ptau Ser202/Thr205 levels correlate in human and transgenic AD mouse models, we analyzed human cortical and hippocampal brain tissue of different Braak stages as well as murine brain tissue of two transgenic mouse models for levels of Aβ-pE(3) and ptau Ser202/Thr205 and correlated the data. Our results show that Aβ-pE(3) formation is increased at early Braak stages while ptau Ser202/Thr205 mostly increases at later stages. Further analyses revealed strongest correlations between the two pathologies in the temporal, frontal, cingulate, and occipital cortex, however correlation in the hippocampus was weaker. Evaluation of murine transgenic brain tissue demonstrated a slow but steady increase of Aβ-pE(3) from 6 to 12 months of age in the cortex and hippocampus of APPSL mice, and a very early and strong Aβ-pE(3) increase in 5xFAD mice. ptau Ser202/Thr205 levels increased at the age of 9 months in APPSL mice and at 6 months in 5xFAD mice. Our results show that Aβ-pE(3) and ptau Ser202/Thr205 levels strongly correlate in human as well as murine tissues, suggesting that tau phosphorylation might be amplified by Aβ-pE(3).
Collapse
Affiliation(s)
| | | | | | - Kerstin Beutl
- QPS Austria GmbH, Grambach, Austria
- FH Joanneum Graz, Graz, Austria
| | | | - Lauren Walker
- Translational and Clinical Research Institute and Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | - Johannes Attems
- Translational and Clinical Research Institute and Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | | |
Collapse
|
14
|
Deike S, Rothemund S, Voigt B, Samantray S, Strodel B, Binder WH. β-Turn mimetic synthetic peptides as amyloid-β aggregation inhibitors. Bioorg Chem 2020; 101:104012. [PMID: 32683138 DOI: 10.1016/j.bioorg.2020.104012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/28/2022]
Abstract
Aggregation of amyloid peptides results in severe neurodegenerative diseases. While the fibril structures of Aβ40 and Aβ42 have been described recently, resolution of the aggregation pathway and evaluation of potent inhibitors still remains elusive, in particular in view of the hairpin-region of Aβ40. We here report the preparation of beta-turn mimetic conjugates containing synthetic turn mimetic structures in the turn region of Aβ40 and Aβ16-35, replacing 2 amino acids in the turn-region G25 - K28. The structure of the turn mimic induces both, acceleration of fibrillation and the complete inhibition of fibrillation, confirming the importance of the turn region on the aggregation. Replacing position G25-S26 provided the best inhibition effect for both beta-turn mimetics, the bicyclic BTD 1 and the aromatic TAA 2, while positions N27-K28 and V24-G25 showed only weaker or no inhibitory effects. When comparing different turn mimetics at the same position (G25-S26), conjugate 1a bearing the BTD turn showed the best inhibition of Aβ40 aggregation, while 5-amino-valeric acid 4a showed the weakest effect. Thus there is a pronounced impact on fibrillation with the chemical nature of the embedded beta-turn-mimic: the conformationally constrained turns 1 and 2 lead to a significantly reduced fibrillation, even inhibiting fibrillation of native Aβ40 when added in amounts down to 1/10, whereas the more flexible beta-turn-mimics 4-amino-benzoic acid 3a and 5-amino-valeric acid 4a lead to enhanced fibrillation. Toxicity-testing of the most successful conjugate showed only minor toxicity in cell-viability assays using the N2a cell line. Structural downsizing lead to the short fragment BTD/peptide Aβ16-35 as inhibitor of the aggregation of Aβ40, opening large potential for further small peptide based inhibitors.
Collapse
Affiliation(s)
- Stefanie Deike
- Department of Chemistry, Martin Luther University Halle-Wittenberg, Von-Danckelmann-Platz 4, 06120 Halle, Germany
| | - Sven Rothemund
- Core Unit Peptid-Technologien, University Leipzig, Liebigstr. 21, 04103 Leipzig, Germany
| | - Bruno Voigt
- Department of Physics, Martin Luther University Halle-Wittenberg, Betty-Heimannstrasse 7 4, 06120 Halle, Germany
| | - Suman Samantray
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Birgit Strodel
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52428 Jülich, Germany; Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Wolfgang H Binder
- Department of Chemistry, Martin Luther University Halle-Wittenberg, Von-Danckelmann-Platz 4, 06120 Halle, Germany.
| |
Collapse
|
15
|
Maiolo D, Pizzi A, Gori A, Bergamaschi G, Pigliacelli C, Gazzera L, Consonni A, Baggi F, Moda F, Baldelli Bombelli F, Metrangolo P, Resnati G. Enhanced self-assembly of the 7–12 sequence of amyloid-β peptide by tyrosine bromination. Supramol Chem 2020. [DOI: 10.1080/10610278.2020.1734203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Daniele Maiolo
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico Di Milano, Milano, Italy
| | - Andrea Pizzi
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico Di Milano, Milano, Italy
| | - Alessandro Gori
- Istituto Di Scienze E Tecnologie Chimiche, National Research Council of Italy, Milano, Italy
| | - Greta Bergamaschi
- Istituto Di Scienze E Tecnologie Chimiche, National Research Council of Italy, Milano, Italy
| | - Claudia Pigliacelli
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico Di Milano, Milano, Italy
- Hyber Center of Excellence, Department of Applied Physics, Aalto University, Espoo, Finland
| | - Lara Gazzera
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico Di Milano, Milano, Italy
| | | | - Fulvio Baggi
- Fondazione IRCCS Istituto Neurologico “Carlo Besta”, 20133 Milano, Italy
| | - Fabio Moda
- Fondazione IRCCS Istituto Neurologico “Carlo Besta”, 20133 Milano, Italy
| | - Francesca Baldelli Bombelli
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico Di Milano, Milano, Italy
| | - Pierangelo Metrangolo
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico Di Milano, Milano, Italy
- Hyber Center of Excellence, Department of Applied Physics, Aalto University, Espoo, Finland
| | - Giuseppe Resnati
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico Di Milano, Milano, Italy
| |
Collapse
|
16
|
Hettmann T, Gillies SD, Kleinschmidt M, Piechotta A, Makioka K, Lemere CA, Schilling S, Rahfeld JU, Lues I. Development of the clinical candidate PBD-C06, a humanized pGlu3-Aβ-specific antibody against Alzheimer's disease with reduced complement activation. Sci Rep 2020; 10:3294. [PMID: 32094456 PMCID: PMC7040040 DOI: 10.1038/s41598-020-60319-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/08/2020] [Indexed: 11/09/2022] Open
Abstract
In clinical trials with early Alzheimer's patients, administration of anti-amyloid antibodies reduced amyloid deposits, suggesting that immunotherapies may be promising disease-modifying interventions against Alzheimer's disease (AD). Specific forms of amyloid beta (Aβ) peptides, for example post-translationally modified Aβ peptides with a pyroglutamate at the N-terminus (pGlu3, pE3), are attractive antibody targets, due to pGlu3-Aβ's neo-epitope character and its propensity to form neurotoxic oligomeric aggregates. We have generated a novel anti-pGlu3-Aβ antibody, PBD-C06, which is based on a murine precursor antibody that binds with high specificity to pGlu3-Aβ monomers, oligomers and fibrils, including mixed aggregates of unmodified Aβ and pGlu3-Aβ peptides. PBD-C06 was generated by first grafting the murine antigen binding sequences onto suitable human variable light and heavy chains. Subsequently, the humanized antibody was de-immunized and site-specific mutations were introduced to restore original target binding, to eliminate complement activation and to improve protein stability. PBD-C06 binds with the same specificity and avidity as its murine precursor antibody and elimination of C1q binding did not compromise Fcγ-receptor binding or in vitro phagocytosis. Thus, PBD-C06 was specifically designed to target neurotoxic aggregates and to avoid complement-mediated inflammatory responses, in order to lower the risk for vasogenic edemas in the clinic.
Collapse
Affiliation(s)
- Thore Hettmann
- Vivoryon Therapeutics AG, Weinbergweg 22, 06120, Halle (Saale), Germany
| | - Stephen D Gillies
- Provenance Biopharmaceuticals, 70 Bedford Rd, Carlisle, MA, 01741, USA
| | - Martin Kleinschmidt
- Fraunhofer Institute for Cell Therapy and Immunology, Department Molecular Drug Biochemistry and Therapy, Weinbergweg 22, 06120, Halle (Saale), Germany
| | - Anke Piechotta
- Fraunhofer Institute for Cell Therapy and Immunology, Department Molecular Drug Biochemistry and Therapy, Weinbergweg 22, 06120, Halle (Saale), Germany
| | - Koki Makioka
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Cynthia A Lemere
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Stephan Schilling
- Vivoryon Therapeutics AG, Weinbergweg 22, 06120, Halle (Saale), Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Department Molecular Drug Biochemistry and Therapy, Weinbergweg 22, 06120, Halle (Saale), Germany
| | - Jens-Ulrich Rahfeld
- Vivoryon Therapeutics AG, Weinbergweg 22, 06120, Halle (Saale), Germany.
- Fraunhofer Institute for Cell Therapy and Immunology, Department Molecular Drug Biochemistry and Therapy, Weinbergweg 22, 06120, Halle (Saale), Germany.
| | - Inge Lues
- Vivoryon Therapeutics AG, Weinbergweg 22, 06120, Halle (Saale), Germany
| |
Collapse
|
17
|
Maiolo D, Pizzi A, Gori A, Gazzera L, Demitri N, Genoni A, Baggi F, Moda F, Terraneo G, Baldelli Bombelli F, Metrangolo P, Resnati G. Halogenation of the N-Terminus Tyrosine 10 Promotes Supramolecular Stabilization of the Amyloid-β Sequence 7-12. ChemistryOpen 2020; 9:253-260. [PMID: 32110506 PMCID: PMC7041548 DOI: 10.1002/open.201900350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/07/2020] [Indexed: 12/16/2022] Open
Abstract
Here, we demonstrate that introduction of halogen atoms at the tyrosine 10 phenol ring of the DSGYEV sequence derived from the flexible amyloid-β N-terminus, promotes its self-assembly in the solid state. In particular, we report the crystal structures of two halogen-modified sequences, which we found to be stabilized in the solid state by halogen-mediated interactions. The structural study is corroborated by Non-Covalent Interaction (NCI) analysis. Our results prove that selective halogenation of an amino acid enhances the supramolecular organization of otherwise unstructured biologically-relevant sequences. This method may develop as a general strategy for stabilizing highly polymorphic peptide regions.
Collapse
Affiliation(s)
- Daniele Maiolo
- Dept. Chem., Mater., and Chem. Eng. “Giulio Natta”Politecnico di MilanoVia L. Mancinelli 720131MilanoItaly
| | - Andrea Pizzi
- Dept. Chem., Mater., and Chem. Eng. “Giulio Natta”Politecnico di MilanoVia L. Mancinelli 720131MilanoItaly
| | - Alessandro Gori
- Istituto di Scienze e Tecnologie ChimicheNational Research Council of ItalyVia M. Bianco 920131MilanoItaly
| | - Lara Gazzera
- Dept. Chem., Mater., and Chem. Eng. “Giulio Natta”Politecnico di MilanoVia L. Mancinelli 720131MilanoItaly
| | - Nicola Demitri
- Elettra – Sincrotrone TriesteS.S. 14 Km 163.5 in Area Science Park34149Basovizza – TriesteItaly
| | - Alessandro Genoni
- Laboratoire de Physique et Chimie ThéoriquesUniversité de Lorraine and CNRS UMR CNRS 70191 Boulevard Arago57078MetzFrance
| | - Fulvio Baggi
- Fondazione IRCCS Istituto Neurologico “Carlo Besta”Via G. Celoria 1120133MilanItaly
| | - Fabio Moda
- Fondazione IRCCS Istituto Neurologico “Carlo Besta”Via G. Celoria 1120133MilanItaly
| | - Giancarlo Terraneo
- Dept. Chem., Mater., and Chem. Eng. “Giulio Natta”Politecnico di MilanoVia L. Mancinelli 720131MilanoItaly
- Istituto di Scienze e Tecnologie ChimicheNational Research Council of ItalyVia M. Bianco 920131MilanoItaly
| | | | - Pierangelo Metrangolo
- Dept. Chem., Mater., and Chem. Eng. “Giulio Natta”Politecnico di MilanoVia L. Mancinelli 720131MilanoItaly
| | - Giuseppe Resnati
- Dept. Chem., Mater., and Chem. Eng. “Giulio Natta”Politecnico di MilanoVia L. Mancinelli 720131MilanoItaly
| |
Collapse
|
18
|
Cline EN, Bicca MA, Viola KL, Klein WL. The Amyloid-β Oligomer Hypothesis: Beginning of the Third Decade. J Alzheimers Dis 2019; 64:S567-S610. [PMID: 29843241 PMCID: PMC6004937 DOI: 10.3233/jad-179941] [Citation(s) in RCA: 598] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The amyloid-β oligomer (AβO) hypothesis was introduced in 1998. It proposed that the brain damage leading to Alzheimer’s disease (AD) was instigated by soluble, ligand-like AβOs. This hypothesis was based on the discovery that fibril-free synthetic preparations of AβOs were potent CNS neurotoxins that rapidly inhibited long-term potentiation and, with time, caused selective nerve cell death (Lambert et al., 1998). The mechanism was attributed to disrupted signaling involving the tyrosine-protein kinase Fyn, mediated by an unknown toxin receptor. Over 4,000 articles concerning AβOs have been published since then, including more than 400 reviews. AβOs have been shown to accumulate in an AD-dependent manner in human and animal model brain tissue and, experimentally, to impair learning and memory and instigate major facets of AD neuropathology, including tau pathology, synapse deterioration and loss, inflammation, and oxidative damage. As reviewed by Hayden and Teplow in 2013, the AβO hypothesis “has all but supplanted the amyloid cascade.” Despite the emerging understanding of the role played by AβOs in AD pathogenesis, AβOs have not yet received the clinical attention given to amyloid plaques, which have been at the core of major attempts at therapeutics and diagnostics but are no longer regarded as the most pathogenic form of Aβ. However, if the momentum of AβO research continues, particularly efforts to elucidate key aspects of structure, a clear path to a successful disease modifying therapy can be envisioned. Ensuring that lessons learned from recent, late-stage clinical failures are applied appropriately throughout therapeutic development will further enable the likelihood of a successful therapy in the near-term.
Collapse
Affiliation(s)
- Erika N Cline
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Maíra Assunção Bicca
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Kirsten L Viola
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - William L Klein
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
19
|
da S. Hage-Melim LI, Ferreira JV, de Oliveira NK, Correia LC, Almeida MR, Poiani JG, Taft CA, de Paula da Silva CH. The Impact of Natural Compounds on the Treatment of Neurodegenerative Diseases. CURR ORG CHEM 2019. [DOI: 10.2174/1385272823666190327100418] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases (NDDs) are characterized by a progressive deterioration of the motor and/or cognitive function, that are often accompanied by psychiatric disorders, caused by a selective loss of neurons in the central nervous system. Among the NDDs we can mention Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), spinocerebellar ataxia 3 (SCA3), spinal and bulbar muscular atrophy (SBMA) and Creutzfeldt-Jakob disease (CJD). AD and HD are characterized mainly by massive neuronal loss. PD, ALS, SCA3 and SBMA are agerelated diseases which have characteristic motor symptoms. CJD is an NDD caused by prion proteins. With increasing life expectancy, elderly populations tend to have more health problems, such as chronic diseases related to age and disability. Therefore, the development of therapeutic strategies to treat or prevent multiple pathophysiological conditions in the elderly can improve the expectation and quality of life. The attention of researchers has been focused on bioactive natural compounds that represent important resources in the discovery and development of drug candidates against NDDs. In this review, we discuss the pathogenesis, symptoms, potential targets, treatment and natural compounds effective in the treatment of AD, PD, HD, ALS, SCA3, SBMA and CJD.
Collapse
Affiliation(s)
- Lorane I. da S. Hage-Melim
- Laboratorio de Quimica Farmaceutica e Medicinal (PharMedChem), Universidade Federal do Amapa, Macapa, Brazil
| | - Jaderson V. Ferreira
- Laboratorio de Quimica Farmaceutica e Medicinal (PharMedChem), Universidade Federal do Amapa, Macapa, Brazil
| | - Nayana K.S. de Oliveira
- Laboratorio de Quimica Farmaceutica e Medicinal (PharMedChem), Universidade Federal do Amapa, Macapa, Brazil
| | - Lenir C. Correia
- Laboratorio de Quimica Farmaceutica e Medicinal (PharMedChem), Universidade Federal do Amapa, Macapa, Brazil
| | - Marcos R.S. Almeida
- Laboratorio de Quimica Farmaceutica e Medicinal (PharMedChem), Universidade Federal do Amapa, Macapa, Brazil
| | - João G.C. Poiani
- Laboratorio Computacional de Química Farmaceutica, Departamento de Ciencias Farmaceuticas, Faculdade de Ciencias Farmaceuticas de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Carlton A. Taft
- Centro Brasileiro de Pesquisas Fisicas, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos H.T. de Paula da Silva
- Laboratorio Computacional de Química Farmaceutica, Departamento de Ciencias Farmaceuticas, Faculdade de Ciencias Farmaceuticas de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
20
|
Wani A, Gupta M, Ahmad M, Shah AM, Ahsan AU, Qazi PH, Malik F, Singh G, Sharma PR, Kaddoumi A, Bharate SB, Vishwakarma RA, Kumar A. Alborixin clears amyloid-β by inducing autophagy through PTEN-mediated inhibition of the AKT pathway. Autophagy 2019; 15:1810-1828. [PMID: 30894052 DOI: 10.1080/15548627.2019.1596476] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Imbalance in production and clearance of amyloid beta (Aβ) is the primary reason for its deposition in Alzheimer disease. Macroautophagy/autophagy is one of the important mechanisms for clearance of both intracellular and extracellular Aβ. Here, through screening, we identified alborixin, an ionophore, as a potent inducer of autophagy. We found that autophagy induced by alborixin substantially cleared Aβ in microglia and primary neuronal cells. Induction of autophagy was accompanied by up regulation of autophagy proteins BECN1/Beclin 1, ATG5, ATG7 and increased lysosomal activities. Autophagy induced by alborixin was associated with inhibition of the phosphoinositide 3-kinase (PI3K)-AKT pathway. A knock down of PTEN and consistent, constitutive activation of AKT inhibited alborixin-induced autophagy and consequent clearance of Aβ. Furthermore, clearance of Aβ by alborixin led to significant reduction of Aβ-mediated cytotoxicity in primary neurons and differentiated N2a cells. Thus, our findings put forward alborixin as a potential anti-Alzheimer therapeutic lead. Abbreviations: Aβ: amyloid beta; ALB: alborixin; ATG: autophagy-related; BECN1: beclin 1; DAPI: 4, 6-diamidino-2-phenylindole; DCFH-DA: 2,7-dichlorodihydrofluorescein diacetate; fAβ: fibrillary form of amyloid beta; GFAP: glial fibrillary acidic protein; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MAP2: microtubule-associated protein 2; MTOR: mechanistic target of rapamycin kinase; PTEN: phosphatase and tensin homolog; ROS: reactive oxygen species; SQSTM1: sequestosome 1; TMRE: tetramethylrhodamine, ethyl ester.
Collapse
Affiliation(s)
- Abubakar Wani
- Division of PK-PD-Toxicology and Formulation, CSIR-Indian Institute of Integrative Medicine , Jammu , India.,Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India
| | - Mehak Gupta
- Division of PK-PD-Toxicology and Formulation, CSIR-Indian Institute of Integrative Medicine , Jammu , India.,Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India
| | - Masroor Ahmad
- Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India.,Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine , Jammu , India
| | - Aabid M Shah
- Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India.,Division of Microbial biotechnology, CSIR-Indian Institute of Integrative Medicine , Jammu , India
| | - Aitizaz Ul Ahsan
- Cytogenetics Laboratory, Department of Zoology, Panjab University , Chandigarh , India
| | - Parvaiz H Qazi
- Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India.,Division of Microbial biotechnology, CSIR-Indian Institute of Integrative Medicine , Jammu , India
| | - Fayaz Malik
- Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India.,Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine , Jammu , India
| | - Gurdarshan Singh
- Division of PK-PD-Toxicology and Formulation, CSIR-Indian Institute of Integrative Medicine , Jammu , India.,Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India
| | - Parduman R Sharma
- Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India.,Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine , Jammu , India
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, 720 S. Donahue Dr., Auburn University , Auburn , AL , USA
| | - Sandip B Bharate
- Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India.,Division of Medicinal Chemistry, CSIR-Indian Institute of Integrative Medicine , Jammu , India
| | - Ram A Vishwakarma
- Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India.,Division of Medicinal Chemistry, CSIR-Indian Institute of Integrative Medicine , Jammu , India
| | - Ajay Kumar
- Division of PK-PD-Toxicology and Formulation, CSIR-Indian Institute of Integrative Medicine , Jammu , India.,Academy of Scientific and Innovative Research (AcSIR) , New Delhi , India
| |
Collapse
|
21
|
de Oliveira NKS, Almeida MRS, Pontes FMM, Barcelos MP, de Paula da Silva CHT, Rosa JMC, Cruz RAS, da Silva Hage-Melim LI. Antioxidant Effect of Flavonoids Present in Euterpe oleracea Martius and Neurodegenerative Diseases: A Literature Review. Cent Nerv Syst Agents Med Chem 2019; 19:75-99. [PMID: 31057125 DOI: 10.2174/1871524919666190502105855] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/05/2019] [Accepted: 04/11/2019] [Indexed: 06/09/2023]
Abstract
INTRODUCTION Neurodegenerative diseases (NDDs) are progressive, directly affecting the central nervous system (CNS), the most common and recurrent are Alzheimer's disease (AD) and Parkinson's disease (PD). One factor frequently mentioned in the etiology of NDDs is the generation of free radicals and oxidative stress, producing cellular damages. Studies have shown that the consumption of foods rich in polyphenols, especially those of the flavonoid class, has been related to the low risk in the development of several diseases. Due to the antioxidant properties present in the food, a fruit that has been gaining prominence among these foods is the Euterpe oleracea Mart. (açaí), because it presents in its composition significant amounts of a subclass of the flavonoids, the anthocyanins. METHODS In the case review, the authors receive a basic background on the most common NDDs, oxidative stress and antioxidants. In addition, revisiting the various studies related to NDDs, including flavonoids and consumption of açaí. RESULTS Detailed analysis of the recently reported case studies reveal that dietary consumption of flavonoid-rich foods, such as açaí fruits, suggests the efficacy to attenuate neurodegeneration and prevent or reverse the age-dependent deterioration of cognitive function. CONCLUSION This systematic review points out that flavonoids presenting in açaí have the potential for the treatment of diseases such as PD and AD and are candidates for drugs in future clinical research. However, there is a need for in vitro and in vivo studies with polyphenol that prove and ratify the therapeutic potential of this fruit for several NDDs.
Collapse
Affiliation(s)
| | - Marcos Rafael Silva Almeida
- Laboratory of Pharmaceutical and Medicinal Chemistry (PharMedChem), Federal University of Amapa, Macapa, Brazil
| | - Franco Márcio Maciel Pontes
- Laboratory of Pharmaceutical and Medicinal Chemistry (PharMedChem), Federal University of Amapa, Macapa, Brazil
| | - Mariana Pegrucci Barcelos
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Carlos Henrique Tomich de Paula da Silva
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Joaquín María Campos Rosa
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
- Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, 18071 Granada, Spain, Instituto de Investigación, Biosanitaria ibs, Granada, Universidad de Granada, Granada, Spain
| | | | | |
Collapse
|
22
|
Pagano K, Galante D, D'Arrigo C, Corsaro A, Nizzari M, Florio T, Molinari H, Tomaselli S, Ragona L. Effects of Prion Protein on Aβ42 and Pyroglutamate-Modified AβpΕ3-42 Oligomerization and Toxicity. Mol Neurobiol 2018; 56:1957-1971. [PMID: 29981054 DOI: 10.1007/s12035-018-1202-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/26/2018] [Indexed: 11/24/2022]
Abstract
Soluble Aβ oligomers are widely recognized as the toxic forms responsible for triggering AD, and Aβ receptors are hypothesized to represent the first step in a neuronal cascade leading to dementia. Cellular prion protein (PrP) has been reported as a high-affinity binder of Aβ oligomers. The interactions of PrP with both Aβ42 and the highly toxic N-truncated pyroglutamylated species (AβpE3-42) are here investigated, at a molecular level, by means of ThT fluorescence, NMR and TEM. We demonstrate that soluble PrP binds both Aβ42 and AβpE3-42, preferentially interacting with oligomeric species and delaying fibril formation. Residue level analysis of Aβ42 oligomerization process reveals, for the first time, that PrP is able to differently interact with the forming oligomers, depending on the aggregation state of the starting Aβ42 sample. A distinct behavior is observed for Aβ42 1-30 region and C-terminal residues, suggesting that PrP protects Aβ42 N-tail from entangling on the mature NMR-invisible fibril, consistent with the hypothesis that Aβ42 N-tail is the locus of interaction with PrP. PrP/AβpE3-42 interactions are here reported for the first time. All interaction data are validated and complemented by cellular tests performed on Wt and PrP-silenced neuronal cell lines, clearly showing PrP dependent Aβ oligomer cell internalization and toxicity. The ability of soluble PrP to compete with membrane-anchored PrP for binding to Aβ oligomers bears relevance for studies of druggable pathways.
Collapse
Affiliation(s)
- Katiuscia Pagano
- Istituto per lo Studio delle Macromolecole (ISMAC), CNR, Milan, Italy
| | | | | | - Alessandro Corsaro
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical research (CEBR), University of Genoa, Genoa, Italy
| | - Mario Nizzari
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical research (CEBR), University of Genoa, Genoa, Italy
| | - Tullio Florio
- Section of Pharmacology, Department of Internal Medicine, and Center of Excellence for Biomedical research (CEBR), University of Genoa, Genoa, Italy
| | | | - Simona Tomaselli
- Istituto per lo Studio delle Macromolecole (ISMAC), CNR, Milan, Italy.
| | - Laura Ragona
- Istituto per lo Studio delle Macromolecole (ISMAC), CNR, Milan, Italy.
| |
Collapse
|
23
|
Mehta PD, Patrick BA, Barshatzky M, Mehta SP, Frackowiak J, Mazur-Kolecka B, Wegiel J, Wisniewski T, Miller DL. Generation and Partial Characterization of Rabbit Monoclonal Antibody to Pyroglutamate Amyloid-β3-42 (pE3-Aβ). J Alzheimers Dis 2018; 62:1635-1649. [DOI: 10.3233/jad-170898] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Pankaj D. Mehta
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Bruce A. Patrick
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Marc Barshatzky
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Sangita P. Mehta
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Janusz Frackowiak
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Bozena Mazur-Kolecka
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Jerzy Wegiel
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology, New York University School of Medicine, New York, NY, USA
| | - David L. Miller
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
24
|
Yano Y, Takeno A, Matsuzaki K. Trace amounts of pyroglutaminated Aβ-(3-42) enhance aggregation of Aβ-(1-42) on neuronal membranes at physiological concentrations: FCS analysis of cell surface. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1603-1608. [PMID: 29410161 DOI: 10.1016/j.bbamem.2018.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/24/2018] [Accepted: 01/27/2018] [Indexed: 10/18/2022]
Abstract
Minor species of amyloid β-peptide (Aβ), such as Aβ-(1-43) and pyroglutaminated Aβ-(3-42) (Aβ-(3pE-42)), have been suggested to be involved in the initiation of the Aβ aggregation process, which is closely associated with the etiology of Alzheimer's disease. They can play important roles in aggregation not only in the aqueous phase but also on neuroral membranes; however, the latter behaviors remain mostly unexplored. Here, initial aggregation processes of Aβ on living cells were monitored at physiological nanomolar concentrations by fluorescence correlation spectroscopy. Membrane-bound Aβ-(1-42) and Aβ-(1-40) formed oligomers composed of ~4 Aβ molecules during 48-h incubation, whereas the peptides remained monomeric in the culture medium, indicating that the membranes facilitated Aβ aggregation. The presence of 5 mol% Aβ-(3pE-42), but not Aβ-(1-43), significantly enhanced the aggregation of Aβ-(1-42) up to ~10-mers. On the other hand, neither trace amounts of Aβ-(1-42) nor Aβ-(3pE-42) enhanced the aggregation of Aβ-(1-40). The observed small Aβ oligomers are expected to act as pathogenic seeds for amyloid fibrils responsible for neurotoxicity. This article is part of a Special Issue entitled: Protein Aggregation and Misfolding at the Cell Membrane Interface edited by Ayyalusamy Ramamoorthy.
Collapse
Affiliation(s)
- Yoshiaki Yano
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - An Takeno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Katsumi Matsuzaki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|