1
|
Shim KH, Sharma N, An SSA. Prion therapeutics: Lessons from the past. Prion 2022; 16:265-294. [PMID: 36515657 PMCID: PMC9754114 DOI: 10.1080/19336896.2022.2153551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 12/15/2022] Open
Abstract
Prion diseases are a group of incurable zoonotic neurodegenerative diseases (NDDs) in humans and other animals caused by the prion proteins. The abnormal folding and aggregation of the soluble cellular prion proteins (PrPC) into scrapie isoform (PrPSc) in the Central nervous system (CNS) resulted in brain damage and other neurological symptoms. Different therapeutic approaches, including stalling PrPC to PrPSc conversion, increasing PrPSc removal, and PrPC stabilization, for which a spectrum of compounds, ranging from organic compounds to antibodies, have been explored. Additionally, a non-PrP targeted drug strategy using serpin inhibitors has been discussed. Despite numerous scaffolds being screened for anti-prion activity in vitro, only a few were effective in vivo and unfortunately, almost none of them proved effective in the clinical studies, most likely due to toxicity and lack of permeability. Recently, encouraging results from a prion-protein monoclonal antibody, PRN100, were presented in the first human trial on CJD patients, which gives a hope for better future for the discovery of other new molecules to treat prion diseases. In this comprehensive review, we have re-visited the history and discussed various classes of anti-prion agents, their structure, mode of action, and toxicity. Understanding pathogenesis would be vital for developing future treatments for prion diseases. Based on the outcomes of existing therapies, new anti-prion agents could be identified/synthesized/designed with reduced toxicity and increased bioavailability, which could probably be effective in treating prion diseases.
Collapse
Affiliation(s)
- Kyu Hwan Shim
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Niti Sharma
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| |
Collapse
|
2
|
The Association of Lipids with Amyloid Fibrils. J Biol Chem 2022; 298:102108. [PMID: 35688209 PMCID: PMC9293637 DOI: 10.1016/j.jbc.2022.102108] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 01/02/2023] Open
Abstract
Amyloid formation continues to be a widely studied area because of its association with numerous diseases, such as Alzheimer’s and Parkinson’s diseases. Despite a large body of work on protein aggregation and fibril formation, there are still significant gaps in our understanding of the factors that differentiate toxic amyloid formation in vivo from alternative misfolding pathways. In addition to proteins, amyloid fibrils are often associated in their cellular context with several types of molecule, including carbohydrates, polyanions, and lipids. This review focuses in particular on evidence for the presence of lipids in amyloid fibrils and the routes by which those lipids may become incorporated. Chemical analyses of fibril composition, combined with studies to probe the lipid distribution around fibrils, provide evidence that in some cases, lipids have a strong association with fibrils. In addition, amyloid fibrils formed in the presence of lipids have distinct morphologies and material properties. It is argued that lipids are an integral part of many amyloid deposits in vivo, where their presence has the potential to influence the nucleation, morphology, and mechanical properties of fibrils. The role of lipids in these structures is therefore worthy of further study.
Collapse
|
3
|
Gupta A, Dey S, Bhowmik D, Maiti S. Coexisting Ordered and Disordered Membrane Phases Have Distinct Modes of Interaction with Disease-Associated Oligomers. J Phys Chem B 2022; 126:1016-1023. [PMID: 35104126 DOI: 10.1021/acs.jpcb.1c09421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ordered membrane domains are thought to influence the attachment and insertion of toxic amyloid oligomers, and consequently, their toxicity. However, if and how the molecular aspects of this interaction depend on the membrane order is poorly understood. Here we measure the affinity, location, and degree of insertion of the small oligomers of hIAPP (human Islet Amyloid Polypeptide, associated with Type II diabetes) at near-physiological concentrations to adjacent domains of a biphasic lipid bilayer. Using simultaneous atomic force, confocal and fluorescence lifetime microscopy (AFM-FLIM), we find that hIAPP oligomers have a nearly 8-fold higher affinity to the disordered domains over the ordered domains. To probe whether this difference indicates different modes of interaction, we measure the change of lifetime of peptide-attached fluorescent labels induced by soluble fluorescence quenchers and also measure the kinetics of localized photobleaching. We find that in the raft-like ordered domains, the oligomers primarily lie on the aqueous interface with limited membrane penetration. However, in the neighboring disordered domains, their C-termini penetrate deeper into the lipid bilayer. We conclude that local membrane order determines not only the affinity but also the mode of interaction of amyloid oligomers, which may have significant implications for disease mechanisms.
Collapse
Affiliation(s)
- Ankur Gupta
- Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai 400005, India
| | - Simli Dey
- Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai 400005, India
| | - Debanjan Bhowmik
- Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai 400005, India
| | - Sudipta Maiti
- Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai 400005, India
| |
Collapse
|
4
|
Crestini A, Santilli F, Martellucci S, Carbone E, Sorice M, Piscopo P, Mattei V. Prions and Neurodegenerative Diseases: A Focus on Alzheimer's Disease. J Alzheimers Dis 2021; 85:503-518. [PMID: 34864675 DOI: 10.3233/jad-215171] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Specific protein misfolding and aggregation are mechanisms underlying various neurodegenerative diseases such as prion disease and Alzheimer's disease (AD). The misfolded proteins are involved in prions, amyloid-β (Aβ), tau, and α-synuclein disorders; they share common structural, biological, and biochemical characteristics, as well as similar mechanisms of aggregation and self-propagation. Pathological features of AD include the appearance of plaques consisting of deposition of protein Aβ and neurofibrillary tangles formed by the hyperphosphorylated tau protein. Although it is not clear how protein aggregation leads to AD, we are learning that the cellular prion protein (PrPC) plays an important role in the pathogenesis of AD. Herein, we first examined the pathogenesis of prion and AD with a focus on the contribution of PrPC to the development of AD. We analyzed the mechanisms that lead to the formation of a high affinity bond between Aβ oligomers (AβOs) and PrPC. Also, we studied the role of PrPC as an AβO receptor that initiates an AβO-induced signal cascade involving mGluR5, Fyn, Pyk2, and eEF2K linking Aβ and tau pathologies, resulting in the death of neurons in the central nervous system. Finally, we have described how the PrPC-AβOs interaction can be used as a new potential therapeutic target for the treatment of PrPC-dependent AD.
Collapse
Affiliation(s)
- Alessio Crestini
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Santilli
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Rieti, Italy.,Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| | - Stefano Martellucci
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Rieti, Italy
| | - Elena Carbone
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| | - Paola Piscopo
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Rieti, Italy.,Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| |
Collapse
|
5
|
The prion fragment PrP106-127 adopts a secondary structure typical of aggregated fibrils in langmuir monolayers of brain lipid extract. Chem Phys Lipids 2020; 230:104930. [DOI: 10.1016/j.chemphyslip.2020.104930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/24/2020] [Accepted: 05/16/2020] [Indexed: 02/07/2023]
|
6
|
Kara S, Amon L, Lühr JJ, Nimmerjahn F, Dudziak D, Lux A. Impact of Plasma Membrane Domains on IgG Fc Receptor Function. Front Immunol 2020; 11:1320. [PMID: 32714325 PMCID: PMC7344230 DOI: 10.3389/fimmu.2020.01320] [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: 03/12/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022] Open
Abstract
Lipid cell membranes not only represent the physical boundaries of cells. They also actively participate in many cellular processes. This contribution is facilitated by highly complex mixtures of different lipids and incorporation of various membrane proteins. One group of membrane-associated receptors are Fc receptors (FcRs). These cell-surface receptors are crucial for the activity of most immune cells as they bind immunoglobulins such as immunoglobulin G (IgG). Based on distinct mechanisms of IgG binding, two classes of Fc receptors are now recognized: the canonical type I FcγRs and select C-type lectin receptors newly referred to as type II FcRs. Upon IgG immune complex induced cross-linking, these receptors are known to induce a multitude of cellular effector responses in a cell-type dependent manner, including internalization, antigen processing, and presentation as well as production of cytokines. The response is also determined by specific intracellular signaling domains, allowing FcRs to either positively or negatively modulate immune cell activity. Expression of cell-type specific combinations and numbers of receptors therefore ultimately sets a threshold for induction of effector responses. Mechanistically, receptor cross-linking and localization to lipid rafts, i.e., organized membrane microdomains enriched in intracellular signaling proteins, were proposed as major determinants of initial FcR activation. Given that immune cell membranes might also vary in their lipid compositions, it is reasonable to speculate, that the cell membrane and especially lipid rafts serve as an additional regulator of FcR activity. In this article, we aim to summarize the current knowledge on the interplay of lipid rafts and IgG binding FcRs with a focus on the plasma membrane composition and receptor localization in immune cells, the proposed mechanisms underlying this localization and consequences for FcR function with respect to their immunoregulatory capacity.
Collapse
Affiliation(s)
- Sibel Kara
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jennifer J Lühr
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Division of Nano-Optics, Max-Planck Institute for the Science of Light, Erlangen, Germany
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.,Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Anja Lux
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
7
|
Agarwal A, Das D, Banerjee T, Mukhopadhyay S. Energy migration captures membrane-induced oligomerization of the prion protein. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140324. [DOI: 10.1016/j.bbapap.2019.140324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/23/2019] [Accepted: 10/26/2019] [Indexed: 12/22/2022]
|
8
|
Ning L, Mu Y. Aggregation of PrP106-126 on surfaces of neutral and negatively charged membranes studied by molecular dynamics simulations. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1936-1948. [PMID: 29550288 DOI: 10.1016/j.bbamem.2018.03.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/08/2018] [Accepted: 03/08/2018] [Indexed: 01/28/2023]
Abstract
Prion diseases are neurodegenerative disorders characterized by the aggregation of an abnormal form of prion protein. The interaction of prion protein and cellular membrane is crucial to elucidate the occurrence and development of prion diseases. Its fragment, residues 106-126, has been proven to maintain the pathological properties of misfolded prion and was used as a model peptide. In this study, explicit solvent molecular dynamics (MD) simulations were carried out to investigate the adsorption, folding and aggregation of PrP106-126 with different sizes (2-peptides, 4-peptides and 6-peptides) on the surface of both pure neutral POPC (1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine) and negatively charged POPC/POPG (1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol) (3:1) lipids. MD simulation results show that PrP106-126 display strong affinity with POPC/POPG but does not interact with pure POPC. The positively charged and polar residues participating hydrogen bonding with membrane promote the adsorption of PrP106-126. The presence of POPC and POPC/POPG exert limited influence on the secondary structures of PrP106-126 and random coil structures are predominant in all simulation systems. Upon the adsorption on the POPC/POPG surface, the aggregation states of PrP106-126 have been changed and more small oligomers were observed. This work provides insights into the interactions of PrP106-126 and membranes with different compositions in atomic level, which expand our understanding the role membrane plays in the development of prion diseases. 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)
- Lulu Ning
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
9
|
Perrier V, Imberdis T, Lafon PA, Cefis M, Wang Y, Huetter E, Arnaud JD, Alvarez-Martinez T, Le Guern N, Maquart G, Lagrost L, Desrumaux C. Plasma cholesterol level determines in vivo prion propagation. J Lipid Res 2017; 58:1950-1961. [PMID: 28765208 PMCID: PMC5625119 DOI: 10.1194/jlr.m073718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 07/28/2017] [Indexed: 12/27/2022] Open
Abstract
Transmissible spongiform encephalopathies are fatal neurodegenerative diseases with an urgent need for therapeutic and prophylactic strategies. At the time when the blood-mediated transmission of prions was demonstrated, in vitro studies indicated a high binding affinity of the scrapie prion protein (PrPSc) with apoB-containing lipoproteins, i.e., the main carriers of cholesterol in human blood. The aim of the present study was to explore the relationship between circulating cholesterol-containing lipoproteins and the pathogenicity of prions in vivo. We showed that, in mice with a genetically engineered deficiency for the plasma lipid transporter, phospholipid transfer protein (PLTP), abnormally low circulating cholesterol concentrations were associated with a significant prolongation of survival time after intraperitoneal inoculation of the 22L prion strain. Moreover, when circulating cholesterol levels rose after feeding PLTP-deficient mice a lipid-enriched diet, a significant reduction in survival time of mice together with a marked increase in the accumulation rate of PrPSc deposits in their brain were observed. Our results suggest that the circulating cholesterol level is a determinant of prion propagation in vivo and that cholesterol-lowering strategies might be a successful therapeutic approach for patients suffering from prion diseases.
Collapse
Affiliation(s)
- Véronique Perrier
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France
| | - Thibaud Imberdis
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France
| | - Pierre-André Lafon
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France
| | - Marina Cefis
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France
| | - Yunyun Wang
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France.,Cellular Signaling Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Elisabeth Huetter
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France
| | - Jacques-Damien Arnaud
- Etablissement Confiné d'Expérimentation A3/L3, CECEMA, US009 Biocampus, UMS 3426, Université Montpellier, Montpellier, F-34095 France
| | - Teresa Alvarez-Martinez
- Etablissement Confiné d'Expérimentation A3/L3, CECEMA, US009 Biocampus, UMS 3426, Université Montpellier, Montpellier, F-34095 France
| | - Naig Le Guern
- INSERM, LNC UMR866, F-21000 Dijon, France and LNC UMR866, Université Bourgogne Franche-Comté, F-21000 Dijon, France.,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, F-21000 Dijon, France
| | - Guillaume Maquart
- INSERM, LNC UMR866, F-21000 Dijon, France and LNC UMR866, Université Bourgogne Franche-Comté, F-21000 Dijon, France.,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, F-21000 Dijon, France
| | - Laurent Lagrost
- INSERM, LNC UMR866, F-21000 Dijon, France and LNC UMR866, Université Bourgogne Franche-Comté, F-21000 Dijon, France.,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, F-21000 Dijon, France.,University Hospital of Dijon, F-21000 Dijon, France
| | - Catherine Desrumaux
- Université Montpellier and Inserm U1198, Montpellier, F-34095 France and EPHE, Paris, F-75007 France .,LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, F-21000 Dijon, France
| |
Collapse
|
10
|
Pan J, Sahoo PK, Dalzini A, Hayati Z, Aryal CM, Teng P, Cai J, Gutierrez HR, Song L. Membrane Disruption Mechanism of a Prion Peptide (106-126) Investigated by Atomic Force Microscopy, Raman and Electron Paramagnetic Resonance Spectroscopy. J Phys Chem B 2017; 121:5058-5071. [PMID: 28459565 PMCID: PMC5770145 DOI: 10.1021/acs.jpcb.7b02772] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A fragment of the human prion protein spanning residues 106-126 (PrP106-126) recapitulates many essential properties of the disease-causing protein such as amyloidogenicity and cytotoxicity. PrP106-126 has an amphipathic characteristic that resembles many antimicrobial peptides (AMPs). Therefore, the toxic effect of PrP106-126 could arise from a direct association of monomeric peptides with the membrane matrix. Several experimental approaches are employed to scrutinize the impacts of monomeric PrP106-126 on model lipid membranes. Porous defects in planar bilayers are observed by using solution atomic force microscopy. Adding cholesterol does not impede defect formation. A force spectroscopy experiment shows that PrP106-126 reduces Young's modulus of planar lipid bilayers. We use Raman microspectroscopy to study the effect of PrP106-126 on lipid atomic vibrational dynamics. For phosphatidylcholine lipids, PrP106-126 disorders the intrachain conformation, while the interchain interaction is not altered; for phosphatidylethanolamine lipids, PrP106-126 increases the interchain interaction, while the intrachain conformational order remains similar. We explain the observed differences by considering different modes of peptide insertion. Finally, electron paramagnetic resonance spectroscopy shows that PrP106-126 progressively decreases the orientational order of lipid acyl chains in magnetically aligned bicelles. Together, our experimental data support the proposition that monomeric PrP106-126 can disrupt lipid membranes by using similar mechanisms found in AMPs.
Collapse
Affiliation(s)
- Jianjun Pan
- Department of Physics, University of South Florida, Tampa, Florida 33620, United States
| | - Prasana K. Sahoo
- Department of Physics, University of South Florida, Tampa, Florida 33620, United States
| | - Annalisa Dalzini
- National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida 32310, United States
| | - Zahra Hayati
- National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida 32310, United States
| | - Chinta M. Aryal
- Department of Physics, University of South Florida, Tampa, Florida 33620, United States
| | - Peng Teng
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | | | - Likai Song
- National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida 32310, United States
| |
Collapse
|
11
|
Dall'Ara P, Iulini B, Botto L, Filipe J, Martino PA, Pintore MD, Gazzuola P, Mazza M, Dagrada M, Ingravalle F, Casalone C, Palestini P, Poli G. Diets with different lipid contents do not modify the neuronal membrane lipid raft profile in a scrapie murine model. Life Sci 2016; 144:226-33. [PMID: 26655166 DOI: 10.1016/j.lfs.2015.12.008] [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: 04/17/2015] [Revised: 11/26/2015] [Accepted: 12/02/2015] [Indexed: 10/22/2022]
Abstract
UNLABELLED In Transmissible Spongiform Encephalopathies (TSEs), the localization of the prion protein in the neuronal membrane lipid rafts (LR) seems to play a role in sustaining the protein misfolding. Changes in membrane properties, due to altered lipid composition, affect their organization and interaction between lipids and protein therein, and consequently also membrane resident protein functionality; dietary polyunsaturated fatty acids (PUFAs), gangliosides and cholesterol seem to influence these processes. AIMS In this work, the influence of administration of different feed, able to change the composition of lipid membrane, on the clinical progression of prion disease was studied. MAIN METHODS The activity of three diets (hyperlipidic with 6% fats; hypolipidic with 0.1% fats; and purified with 4% fats) was tested in CD1 mouse model experimentally infected with RML scrapie strain. Presence and distribution of typical central nervous system (CNS) lesions and deposits of PrP(sc) were evaluated by histopathological analysis and immunohistochemistry. Analysis of lipids was performed in homogenate and insoluble brain fraction of the neuronal membrane rich in LR. KEY FINDINGS Results show that a diet with a different lipid level has not a significant role in the development of the scrapie disease. All infected mice fed with different diets died in the same time span. Histology, immunohistochemistry, and neuropathological analyses of the infected brains did not show significant differences between animals subjected to different diets. SIGNIFICANCE Independently of the diet, the infection induced a significant modification of the lipid composition in homogenates, and a less noticeable one in insoluble brain fraction.
Collapse
Affiliation(s)
- Paola Dall'Ara
- Department of Veterinary Science and Public Health, University of Milan, Milan, Italy.
| | - Barbara Iulini
- CEA, Italian Reference Laboratory for TSEs, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Turin, Italy
| | - Laura Botto
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Joel Filipe
- Department of Veterinary Science and Public Health, University of Milan, Milan, Italy
| | - Piera Anna Martino
- Department of Veterinary Science and Public Health, University of Milan, Milan, Italy
| | - Maria Domenica Pintore
- CEA, Italian Reference Laboratory for TSEs, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Turin, Italy
| | - Paola Gazzuola
- CEA, Italian Reference Laboratory for TSEs, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Turin, Italy
| | - Maria Mazza
- CEA, Italian Reference Laboratory for TSEs, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Turin, Italy
| | - Massimo Dagrada
- Department of Veterinary Science and Public Health, University of Milan, Milan, Italy
| | - Francesco Ingravalle
- CEA, Italian Reference Laboratory for TSEs, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Turin, Italy
| | - Cristina Casalone
- CEA, Italian Reference Laboratory for TSEs, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Turin, Italy
| | - Paola Palestini
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Giorgio Poli
- Department of Veterinary Science and Public Health, University of Milan, Milan, Italy
| |
Collapse
|
12
|
Conformational Switching and Nanoscale Assembly of Human Prion Protein into Polymorphic Amyloids via Structurally Labile Oligomers. Biochemistry 2015; 54:7505-13. [DOI: 10.1021/acs.biochem.5b01110] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
13
|
Contrasting Effects of Two Lipid Cofactors of Prion Replication on the Conformation of the Prion Protein. PLoS One 2015; 10:e0130283. [PMID: 26090881 PMCID: PMC4474664 DOI: 10.1371/journal.pone.0130283] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 05/18/2015] [Indexed: 01/03/2023] Open
Abstract
Recent studies introduced two experimental protocols for converting full-length recombinant prion protein (rPrP) purified from E.coli into the infectious prion state (PrPSc) with high infectivity titers. Both protocols employed protein misfolding cyclic amplification (PMCA) for generating PrPScde novo, but used two different lipids, 1-palmitoyl-2-oleolyl-sn-glycero-3-phospho(1’-rac-glycerol) (POPG) or phosphatidylethanolamine (PE), as conversion cofactors. The current study compares the effect of POPG and PE on the physical properties of native, α-helical full-length mouse rPrP under the solvent conditions used for converting rPrP into PrPSc. Surprisingly, the effects of POPG and PE on rPrP physical properties, including its conformation, thermodynamic stability, aggregation state and interaction with a lipid, were found to be remarkably different. PE was shown to have minimal, if any, effects on rPrP thermodynamic stability, cooperativity of unfolding, immediate solvent environment or aggregation state. In fact, little evidence indicates that PE interacts with rPrP directly. In contrast, POPG was found to bind to and induce dramatic changes in rPrP structure, including a loss of α-helical conformation and formation of large lipid-protein aggregates that were resistant to partially denaturing conditions. These results suggest that the mechanisms by which lipids assist conversion of rPrP into PrPSc might be fundamentally different for POPG and PE.
Collapse
|
14
|
Prion protein misfolding, strains, and neurotoxicity: an update from studies on Mammalian prions. Int J Cell Biol 2013; 2013:910314. [PMID: 24454379 PMCID: PMC3884631 DOI: 10.1155/2013/910314] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 11/10/2013] [Accepted: 11/11/2013] [Indexed: 11/17/2022] Open
Abstract
Prion diseases, also known as transmissible spongiform encephalopathies (TSEs), are a group of fatal neurodegenerative disorders affecting humans and other mammalian species. The central event in TSE pathogenesis is the conformational conversion of the cellular prion protein, PrPC, into the aggregate, β-sheet rich, amyloidogenic form, PrPSc. Increasing evidence indicates that distinct PrPSc conformers, forming distinct ordered aggregates, can encipher the phenotypic TSE variants related to prion strains. Prion strains are TSE isolates that, after inoculation into syngenic hosts, cause disease with distinct characteristics, such as incubation period, pattern of PrPSc distribution, and regional severity of histopathological changes in the brain. In analogy with other amyloid forming proteins, PrPSc toxicity is thought to derive from the existence of various intermediate structures prior to the amyloid fiber formation and/or their specific interaction with membranes. The latter appears particularly relevant for the pathogenesis of TSEs associated with GPI-anchored PrPSc, which involves major cellular membrane distortions in neurons. In this review, we update the current knowledge on the molecular mechanisms underlying three fundamental aspects of the basic biology of prions such as the putative mechanism of prion protein conversion to the pathogenic form PrPSc and its propagation, the molecular basis of prion strains, and the mechanism of induced neurotoxicity by PrPSc aggregates.
Collapse
|
15
|
Rosa A, Scano P, Incani A, Pilla F, Maestrale C, Manca M, Ligios C, Pani A. Lipid profiles in brains from sheep with natural scrapie. Chem Phys Lipids 2013; 175-176:33-40. [DOI: 10.1016/j.chemphyslip.2013.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 08/01/2013] [Indexed: 01/03/2023]
|
16
|
Sanghera N, Correia BEFS, Correia JRS, Ludwig C, Agarwal S, Nakamura HK, Kuwata K, Samain E, Gill AC, Bonev BB, Pinheiro TJT. Deciphering the molecular details for the binding of the prion protein to main ganglioside GM1 of neuronal membranes. ACTA ACUST UNITED AC 2012; 18:1422-31. [PMID: 22118676 DOI: 10.1016/j.chembiol.2011.08.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 08/15/2011] [Accepted: 08/16/2011] [Indexed: 10/15/2022]
Abstract
The prion protein (PrP) resides in lipid rafts in vivo, and lipids modulate misfolding of the protein to infectious isoforms. Here we demonstrate that binding of recombinant PrP to model raft membranes requires the presence of ganglioside GM1. A combination of liquid- and solid-state NMR revealed the binding sites of PrP to the saccharide head group of GM1. The binding epitope for GM1 was mapped to the folded C-terminal domain of PrP, and docking simulations identified key residues in the C-terminal region of helix C and the loop between strand S2 and helix B. Crucially, this region of PrP is linked to prion resistance in vivo, and structural changes caused by lipid binding in this region may explain the requirement for lipids in the generation of infectious prions in vitro.
Collapse
Affiliation(s)
- Narinder Sanghera
- School of Life Sciences, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Butterfield SM, Lashuel HA. Amyloidogenic protein-membrane interactions: mechanistic insight from model systems. Angew Chem Int Ed Engl 2011; 49:5628-54. [PMID: 20623810 DOI: 10.1002/anie.200906670] [Citation(s) in RCA: 483] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The toxicity of amyloid-forming proteins is correlated with their interactions with cell membranes. Binding events between amyloidogenic proteins and membranes result in mutually disruptive structural perturbations, which are associated with toxicity. Membrane surfaces promote the conversion of amyloid-forming proteins into toxic aggregates, and amyloidogenic proteins, in turn, compromise the structural integrity of the cell membrane. Recent studies with artificial model membranes have highlighted the striking resemblance of the mechanisms of membrane permeabilization of amyloid-forming proteins to those of pore-forming toxins and antimicrobial peptides.
Collapse
Affiliation(s)
- Sara M Butterfield
- Laboratory of Molecular Neurobiology and Neuroproteomics, Swiss Federal Institute of Technology Lausanne (EPFL), SV-BMI-LMNN AI2351, 1015 Lausanne, Switzerland
| | | |
Collapse
|
18
|
Evrard SG, Brusco A. Ethanol Effects on the Cytoskeleton of Nerve Tissue Cells. ADVANCES IN NEUROBIOLOGY 2011. [DOI: 10.1007/978-1-4419-6787-9_29] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
19
|
Choi I, Yang YI, Song HD, Lee JS, Kang T, Sung JJ, Yi J. Lipid molecules induce the cytotoxic aggregation of Cu/Zn superoxide dismutase with structurally disordered regions. Biochim Biophys Acta Mol Basis Dis 2010; 1812:41-8. [PMID: 20837142 DOI: 10.1016/j.bbadis.2010.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Revised: 08/18/2010] [Accepted: 09/07/2010] [Indexed: 02/02/2023]
Abstract
Cu/Zn-superoxide dismutase (SOD1) is present in the cytosol, nucleus, peroxisomes and mitochondrial intermembrane space of human cells. More than 114 variants of human SOD1 have been linked to familial amyotrophic lateral sclerosis (ALS), which is also known as Lou Gehrig's disease. Although the ultimate mechanisms underlying SOD1-mediated cytotoxicity are largely unknown, SOD1 aggregates have been strongly implicated as a common feature in ALS. This study examined the mechanism for the formation of SOD1 aggregates in vitro as well as the nature of its cytotoxicity. The aggregation propensity of SOD1 species was investigated using techniques ranging from circular dichroism spectroscopy to fluorescence dye binding methods, as well as electron microscopic imaging. The aggregation of SOD1 appears to be related to its structural instability. The demetallated (apo)-SOD1 and aggregated SOD1 species, with structurally disordered regions, readily undergo aggregation in the presence of lipid molecules, whereas metallated (holo)-SOD1 does not. The majority of aggregated SOD1s that are induced by lipid molecules have an amorphous morphology and exhibit significant cytotoxicity. The lipid binding propensity of SOD1 was found to be closely related to the changes in surface hydrophobicity of the proteins, even at very low levels, which induced further binding and assembly with lipid molecules. These findings suggest that lipid molecules induce SOD1 aggregation under physiological conditions and exert cytotoxicity, and might provide a possible mechanism for the pathogenesis of ALS.
Collapse
Affiliation(s)
- Inhee Choi
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 151-742, Korea
| | | | | | | | | | | | | |
Collapse
|
20
|
Butterfield S, Lashuel H. Wechselwirkungen zwischen amyloidogenen Proteinen und Membranen: Modellsysteme liefern mechanistische Einblicke. Angew Chem Int Ed Engl 2010. [DOI: 10.1002/ange.200906670] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
21
|
Cecarini V, Bonfili L, Cuccioloni M, Mozzicafreddo M, Angeletti M, Eleuteri AM. The relationship between the 20S proteasomes and prion-mediated neurodegenerations: potential therapeutic opportunities. Apoptosis 2010; 15:1322-35. [DOI: 10.1007/s10495-010-0480-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
22
|
Scheinost JC, Witter DP, Boldt GE, Offer J, Wentworth P. Cholesterol secosterol adduction inhibits the misfolding of a mutant prion protein fragment that induces neurodegeneration. Angew Chem Int Ed Engl 2010; 48:9469-72. [PMID: 19899085 DOI: 10.1002/anie.200904524] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Johanna C Scheinost
- The Scripps-Oxford Laboratory, Department of Biochemistry, University of Oxford, South Parks Rd., Oxford OX13QU, UK
| | | | | | | | | |
Collapse
|
23
|
Scheinost J, Witter D, Boldt G, Offer J, Wentworth P. Cholesterol Secosterol Adduction Inhibits the Misfolding of a Mutant Prion Protein Fragment that Induces Neurodegeneration. Angew Chem Int Ed Engl 2009. [DOI: 10.1002/ange.200904524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
24
|
Dorosz J, Volinsky R, Bazar E, Kolusheva S, Jelinek R. Phospholipid-induced fibrillation of a prion amyloidogenic determinant at the air/water interface. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2009; 25:12501-12506. [PMID: 19588938 DOI: 10.1021/la901750v] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The peptide fragment 106-126 of prion protein [PrP(106-126)] is a prominent amyloidogenic determinant. We present analysis of PrP(106-126) fibrillation at the air/water interface and, in particular, the relationship between the fibrillation process and interactions of the peptide with phospholipid monolayers. We find that lipid monolayers deposited at the air/water interface induce rapid formation of remarkably highly ordered fibrils by PrP(106-126), and that the extent of fibrillation and fiber organization were dependent upon the presence of negatively charged and unsaturated phospholipids in the monolayers. We also observe that fibrillation was enhanced when PrP(106-126) was injected underneath preassembled phospholipid monolayers, compared to deposition and subsequent compression of mixed monolayers of the peptide and phospholipids. In a broader context, this study demonstrates that Langmuir systems constitute a useful platform for studying lipid interactions of amyloidogenic peptides and lipid-induced fibrillation phenomena.
Collapse
Affiliation(s)
- Jerzy Dorosz
- Department of Chemistry and Ilse Katz Institute of Nanotechnology, Ben Gurion University, Beer Sheva 84105, Israel
| | | | | | | | | |
Collapse
|
25
|
Copper, iron, and zinc ions homeostasis and their role in neurodegenerative disorders (metal uptake, transport, distribution and regulation). Coord Chem Rev 2009. [DOI: 10.1016/j.ccr.2009.05.011] [Citation(s) in RCA: 342] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
26
|
Shin JY, Shin JI, Kim JS, Yang YS, Shin YK, Kim KK, Lee S, Kweon DH. Disulfide bond as a structural determinant of prion protein membrane insertion. Mol Cells 2009; 27:673-80. [PMID: 19533034 DOI: 10.1007/s10059-009-0089-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 04/21/2009] [Accepted: 04/22/2009] [Indexed: 10/20/2022] Open
Abstract
Conversion of the normal soluble form of prion protein, PrP (PrP(C)), to proteinase K-resistant form (PrP(Sc)) is a common molecular etiology of prion diseases. Proteinase K-resistance is attributed to a drastic conformational change from alpha-helix to beta-sheet and subsequent fibril formation. Compelling evidence suggests that membranes play a role in the conformational conversion of PrP. However, biophysical mechanisms underlying the conformational changes of PrP and membrane binding are still elusive. Recently, we demonstrated that the putative transmembrane domain (TMD; residues 111-135) of Syrian hamster PrP penetrates into the membrane upon the reduction of the conserved disulfide bond of PrP. To understand the mechanism underlying the membrane insertion of the TMD, here we explored changes in conformation and membrane binding abilities of PrP using wild type and cysteine-free mutant. We show that the reduction of the disulfide bond of PrP removes motional restriction of the TMD, which might, in turn, expose the TMD into solvent. The released TMD then penetrates into the membrane. We suggest that the disulfide bond regulates the membrane binding mode of PrP by controlling the motional freedom of the TMD.
Collapse
Affiliation(s)
- Jae Yoon Shin
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Myelin, DIGs, and membrane rafts in the central nervous system. Prostaglandins Other Lipid Mediat 2009; 91:118-29. [PMID: 19379822 DOI: 10.1016/j.prostaglandins.2009.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 04/08/2009] [Accepted: 04/09/2009] [Indexed: 11/21/2022]
Abstract
Over the past 40 years our understanding of the organization of cell membranes has changed dramatically. Membranes are no longer viewed as a homogenous sea of phospholipids studded with randomly positioned islands of proteins. Our current view of the membrane involves the formation of small lipid clusters, comprised mainly of cholesterol and sphingolipids, known as membrane rafts. These lipid clusters apparently include and exclude specific proteins leading to the hypothesis that these domains (1) regulate cellular polarity and compartmentalization through trafficking and sorting, (2) provide platforms for cellular signaling and adhesion, and (3) function as cellular gate keepers. Tremendous controversy surrounds the concept of membrane rafts primarily because these small, highly dynamic entities are too small to be observed with traditional microscopic methods and the most utilized approach for raft analysis relies on poorly quantified, inconsistent biochemical extractions. New analytical approaches are being developed and applied to the study of membrane rafts and these techniques provide great promise for furthering our understanding of these enigmatic domains. In this review we will provide a brief summary of the current understanding of membrane rafts, utilizing the CNS myelin literature for illustrative purposes, and present caveats that should be considered when studying these domains.
Collapse
|
28
|
Electron tomography of early melanosomes: implications for melanogenesis and the generation of fibrillar amyloid sheets. Proc Natl Acad Sci U S A 2008; 105:19726-31. [PMID: 19033461 DOI: 10.1073/pnas.0803488105] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Melanosomes are lysosome-related organelles (LROs) in which melanins are synthesized and stored. Early stage melanosomes are characterized morphologically by intralumenal fibrils upon which melanins are deposited in later stages. The integral membrane protein Pmel17 is a component of the fibrils, can nucleate fibril formation in the absence of other pigment cell-specific proteins, and forms amyloid-like fibrils in vitro. Before fibril formation Pmel17 traffics through multivesicular endosomal compartments, but how these compartments participate in downstream events leading to fibril formation is not fully known. By using high-pressure freezing of MNT-1 melanoma cells and freeze substitution to optimize ultrastructural preservation followed by double tilt 3D electron tomography, we show that the amyloid-like fibrils begin to form in multivesicular compartments, where they radiate from the luminal side of intralumenal membrane vesicles. The fibrils in fully formed stage II premelanosomes organize into sheet-like arrays and exclude the remaining intralumenal vesicles, which are smaller and often in continuity with the limiting membrane. These observations indicate that premelanosome fibrils form in association with intralumenal endosomal membranes. We suggest that similar processes regulate amyloid formation in pathological models.
Collapse
|
29
|
Shin JI, Shin JY, Kim JS, Yang YS, Shin YK, Kweon DH. Deep membrane insertion of prion protein upon reduction of disulfide bond. Biochem Biophys Res Commun 2008; 377:995-1000. [PMID: 18955027 DOI: 10.1016/j.bbrc.2008.10.095] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Accepted: 10/21/2008] [Indexed: 11/24/2022]
Abstract
The membrane may play a role in the pathogenesis of the prion protein (PrP). Cytoplasmic expression of PrP causes the conversion of PrP to a self-perpetuating PrP(Sc)-like conformation and the interaction of polypeptide chain with the hydrophobic core of the membrane is believed to be closely correlated with neurodegeneration. However, it is still elusive what factors govern the membrane interaction of PrP. Here, we show that PrP penetrates deeply into the membrane when the single disulfide bond is reduced, which results in membrane disruption and leakage. The proteinase K treatment and the fluorescence quenching assays showed that a predicted transmembrane domain of PrP penetrates into the membrane when the disulfide bond was reduced. Therefore, the oxidation state of PrP might be an important factor that influences its neurotoxicity or pathogenesis.
Collapse
Affiliation(s)
- Jae-Il Shin
- School of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Gyeonggi-do 440-746, Republic of Korea
| | | | | | | | | | | |
Collapse
|
30
|
Bate C, Tayebi M, Diomede L, Salmona M, Williams A. Docosahexaenoic and eicosapentaenoic acids increase prion formation in neuronal cells. BMC Biol 2008; 6:39. [PMID: 18789130 PMCID: PMC2556658 DOI: 10.1186/1741-7007-6-39] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Accepted: 09/12/2008] [Indexed: 12/12/2022] Open
Abstract
Background The transmissible spongiform encephalopathies, otherwise known as prion diseases, occur following the conversion of the cellular prion protein (PrPC) to an alternatively folded, disease-associated isoform (PrPSc). Recent studies suggest that this conversion occurs via a cholesterol-sensitive process, as cholesterol synthesis inhibitors reduced the formation of PrPSc and delayed the clinical phase of scrapie infection. Since polyunsaturated fatty acids also reduced cellular cholesterol levels we tested their effects on PrPSc formation in three prion-infected neuronal cell lines (ScGT1, ScN2a and SMB cells). Results We report that treatment with docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA) or the cholesterol synthesis inhibitor simvastatin reduced the amounts of free cholesterol in membrane extracts from prion-infected neuronal cells. Simvastatin reduced cholesterol production while DHA and EPA promoted the conversion of free cholesterol to cholesterol esters. Crucially, while simvastatin reduced PrPSc formation, both DHA and EPA significantly increased the amounts of PrPSc in these cells. Unlike simvastatin, the effects of DHA and EPA on PrPSc content were not reversed by stimulation of cholesterol synthesis with mevalonate. Treatment of ScGT1 cells with DHA and EPA also increased activation of cytoplasmic phospholipase A2 and prostaglandin E2 production. Finally, treatment of neuronal cells with DHA and EPA increased the amounts of PrPC expressed at the cell surface and significantly increased the half-life of biotinylated PrPC. Conclusion We report that although treatment with DHA or EPA significantly reduced the free cholesterol content of prion-infected cells they significantly increased PrPSc formation in three neuronal cell lines. DHA or EPA treatment of infected cells increased activation of phospholipase A2, a key enzyme in PrPSc formation, and altered the trafficking of PrPC. PrPC expression at the cell surface, a putative site for the PrPSc formation, was significantly increased, and the rate at which PrPC was degraded was reduced. Cholesterol depletion is seen as a potential therapeutic strategy for prion diseases. However, these results indicate that a greater understanding of the precise relationship between membrane cholesterol distribution, PrPC trafficking, cell activation and PrPSc formation is required before cholesterol manipulation can be considered as a prion therapeutic.
Collapse
Affiliation(s)
- Clive Bate
- Department of Pathology and Infectious Diseases, Royal Veterinary College, Hawkshead Lane, North Mymms, Herts, UK, AL9 7TA.
| | | | | | | | | |
Collapse
|
31
|
Schubert T, Bärmann M, Rusp M, Gränzer W, Tanaka M. Diffusion of glycosylphosphatidylinositol (GPI)-anchored bovine prion protein (PrPc) in supported lipid membranes studied by single-molecule and complementary ensemble methods. J Memb Sci 2008. [DOI: 10.1016/j.memsci.2007.10.058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
32
|
Ultrastructural evidence that ependymal cells are infected in experimental scrapie. Acta Neuropathol 2008; 115:643-50. [PMID: 18369649 DOI: 10.1007/s00401-008-0365-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 03/10/2008] [Accepted: 03/10/2008] [Indexed: 10/22/2022]
Abstract
During the last stage of infection in the experimental scrapie-infected hamster model, light microscopy reveals typical immunostaining of PrPsc in the subependymal region and at the apical ependymal cell borders. Whereas the subependymal immuno-staining is known to originate from extracellular amyloid filaments and residual membranes of astrocytes as constituents of plaque-like structures, the ultrastructural correlate of the supraependymal PrPsc staining remains uncertain. To decipher this apical PrPsc immunopositivity and subsequently the ependymocyte-scrapie agent interaction, we employed highly sensitive immuno-electron microscopy for detecting PrPsc in 263K scrapie-infected hamster brains. The results revealed the supraependymal PrPsc signal to be correlated not only with extracellular accumulation of amyloid filaments, but also with three distinct ependymal cell structures: (1) morphologically intact or altered microvilli associated with filaments, (2) the ependymal cell cytoplasm in proximity of apical cell membrane, and (3) intracytoplasmic organelles such as endosomes and lysosomal-like structures. These findings suggest a strong ependymotrope feature of the scrapie agent and recapitulate several aspects of the cell-prion interaction leading to the formation and production of PrPsc amyloid filaments. Our data demonstrate that in addition to neurons and astrocytes, ependymocytes constitute a new cellular target for the scrapie agent. In contrast, the absence of PrPsc labeling in choroid plexus and brain vascular endothelial cells indicates that these cells are not susceptible to the infection and may inhibit passage of the infectious agent across the blood-brain barrier.
Collapse
|
33
|
Abstract
Transmissible spongiform encephalopathies are neurodegenerative diseases characterized by the accumulation of an abnormal isoform of the prion protein PrP(Sc). Its fragment 106-126 has been reported to maintain most of the pathological features of PrP(Sc), and a role in neurodegeneration has been proposed based on the modulation of membrane properties and channel formation. The ability of PrP(Sc) to modulate membranes and/or form channels in membranes has not been clearly demonstrated; however, if these processes are important, peptide-membrane interactions would be a key feature in the toxicity of PrP(Sc). In this work, the interaction of PrP(106-126) with model membranes comprising typical lipid identities, as well as more specialized lipids such as phosphatidylserine and GM1 ganglioside, was examined using surface plasmon resonance and fluorescence methodologies. This comprehensive study examines different parameters relevant to characterization of peptide-membrane interactions, including membrane charge, viscosity, lipid composition, pH, and ionic strength. We report that PrP(106-126) has a low affinity for lipid membranes under physiological conditions without evidence of membrane disturbances. Membrane insertion and leakage occur only under conditions in which strong electrostatic interactions operate. These results support the hypothesis that the physiological prion protein PrP(C) mediates PrP(106-126) toxic effects in neuronal cells.
Collapse
|
34
|
Schiff E, Campana V, Tivodar S, Lebreton S, Gousset K, Zurzolo C. Coexpression of wild-type and mutant prion proteins alters their cellular localization and partitioning into detergent-resistant membranes. Traffic 2008; 9:1101-15. [PMID: 18410485 DOI: 10.1111/j.1600-0854.2008.00746.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs) are a group of diseases of infectious, sporadic and genetic origin, found in higher organisms and caused by the pathological form of the prion protein. The inheritable subgroup of TSEs is linked to insertional or point mutations in the prion gene prnp, which favour its misfolding and are passed on to offspring in an autosomal-dominant fashion. The large majority of patients with these diseases are heterozygous for the prnp gene, leading to the coexpression of the wild-type (wt) (PrP(C)) and the mutant forms (PrPmut) in the carriers of these mutations. To mimic this situation in vitro, we produced Fischer rat thyroid cells coexpressing PrPwt alongside mutant versions of mouse PrP including A117V, E200K and T182A relevant to the human TSE diseases Gestmann-Sträussler-Scheinker (GSS) disease and familial Creutzfeldt-Jakob disease (fCJD). We found that coexpression of mutant PrP with wt proteins does not affect the glycosylation pattern or the biochemical characteristics of either protein. However, FRET and co-immunoprecipitation experiments suggest an interaction occurring between the wt and mutant proteins. Furthermore, by comparing the intracellular localization and detergent-resistant membrane (DRM) association in single- and double-expressing clones, we found changes in the intracellular/surface ratio and an increased sequestration of both proteins in DRMs, a site believed to be involved in the pathological conversion (or protection thereof) of the prion protein. We, therefore, propose that the mutant forms alter the subcellular localization and the membrane environment of the wt protein in co-transfected cells. These effects may play a role in the development of these diseases.
Collapse
Affiliation(s)
- Edwin Schiff
- Unité de Trafic Membranaire et Pathogénèse, Institut Pasteur, 25 rue du Docteur Roux, 75724 Paris Cedex 15, France
| | | | | | | | | | | |
Collapse
|
35
|
Fournier JG. Cellular prion protein electron microscopy: attempts/limits and clues to a synaptic trait. Implications in neurodegeneration process. Cell Tissue Res 2008; 332:1-11. [PMID: 18236081 DOI: 10.1007/s00441-007-0565-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Accepted: 11/20/2007] [Indexed: 11/24/2022]
Abstract
Prion diseases are caused by an infectious agent constituted by a rogue protein called prion (PrP Sc) of neuronal origin (PrP c) and are exemplified by Creutzfeldt-Jakob disease in humans and bovine spongiform encephalopathy in cattle. Considerable efforts have been made to understand the cerebral damage caused by these diseases but a clear comprehensive view cannot be achieved without defining the neurophysiological function of PrP c. This lack of information is in part attributable to our ignorance of the precise localization of PrP c in the brain neuronal cell. One relevant option to explore this aspect is to undertake PrP immunohistochemistry at the electron-microscopy level, knowing that this challenge raises major technical constraints. In describing the attempts and restrictions of the various approaches used, we review here the efforts that have been invested in this particular field of prionology. The common result emerging from these contributions is that the synapse could be the site at which PrP c exerts its critical activity. This location suggests, in the perspective of synaptic regulation, that PrP c can be assigned multiple biological functions and supports the novel concept that prion-like changes are involved in long-term memory formation. The synaptic trait of PrP c and PrP Sc suggests that synapse loss is the key event in neuronal death. Interestingly, synaptic alterations are also considered to be predominant in the pathophysiological mechanism in Alzheimer, Parkinson and Huntington diseases. All these brain disorders, characterized by the formation of a specific amyloid protein of synaptic origin, can be classified under the heading of amyloidogenic synaptopathies.
Collapse
Affiliation(s)
- Jean-Guy Fournier
- SEPIA/DSV/DRM/CEA, 18 Route Panorama, 92260, Fontenay aux Roses, France.
| |
Collapse
|
36
|
Wegmann S, Miesbauer M, Winklhofer KF, Tatzelt J, Muller DJ. Observing fibrillar assemblies on scrapie-infected cells. Pflugers Arch 2008; 456:83-93. [DOI: 10.1007/s00424-007-0433-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Revised: 12/08/2007] [Accepted: 12/10/2007] [Indexed: 11/29/2022]
|
37
|
Rivaroli A, Prioni S, Loberto N, Bettiga A, Chigorno V, Prinetti A, Sonnino S. Reorganization of prion protein membrane environment during low potassium-induced apoptosis in primary rat cerebellar neurons. J Neurochem 2007; 103:1954-67. [PMID: 17854348 DOI: 10.1111/j.1471-4159.2007.04890.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We studied the changes occurring in the membrane environment of prion protein (PrP) during apoptosis induced by low potassium in primary rat cerebellar neurons. Ceramide levels increased during apoptosis-inducing treatment, being doubled with respect to time-matched controls after 24 h. Sphingomyelin levels were parallely decreased, while cholesterol and ganglioside contents were not affected. Changes in ceramide and sphingomyelin composition were exclusively restricted to a detergent-resistant membrane fraction. The pro-apoptotic treatment was accompanied by the down-regulation of PrP and of the non-receptor kinase Fyn. The levels of PrP and Fyn were correspondingly reduced in the detergent-resistant membrane fraction. In control cells, the membrane microenvironment separated by immunoprecipitation with anti-PrP antibody contained 80% of the detergent-resistant PrP and 35% and 38% of the sphingolipids and cholesterol respectively. Upon low potassium treatment, 20% of the PrP originally present in the detergent-resistant fraction was immunoprecipitated, together with 19% of sphingolipids and 22% of cholesterol. Thus, PrP in the immunoprecipitate from apoptotic cells was ninefold less than in control ones, while sphingolipids and cholesterol were about 50% with respect to controls cells. The molar ratio between cholesterol, sphingomyelin and ceramide was 15 : 6 : 1 in the PrP-rich environment from control neurons, and 6 : 2 : 1 in that from apoptotic cells.
Collapse
Affiliation(s)
- Anna Rivaroli
- Center of Excellence on Neurodegenerative Diseases, Study Center for the Biochemistry and Biotechnology of Glycolipids, Department of Medical Chemistry, Biochemistry and Biotechnology, University of Milan, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
38
|
Stöckl M, Fischer P, Wanker E, Herrmann A. Alpha-synuclein selectively binds to anionic phospholipids embedded in liquid-disordered domains. J Mol Biol 2007; 375:1394-404. [PMID: 18082181 DOI: 10.1016/j.jmb.2007.11.051] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 10/15/2007] [Accepted: 11/15/2007] [Indexed: 11/27/2022]
Abstract
Previous studies indicate that binding of alpha-synuclein to membranes is critical for its physiological function and the development of Parkinson's disease (PD). Here, we have investigated the association of fluorescence-labeled alpha-synuclein variants with different types of giant unilamellar vesicles using confocal microscopy. We found that alpha-synuclein binds with high affinity to anionic phospholipids, when they are embedded in a liquid-disordered as opposed to a liquid-ordered environment. This indicates that not only electrostatic forces but also lipid packing and hydrophobic interactions are critical for the association of alpha-synuclein with membranes in vitro. When compared to wild-type alpha-synuclein, the disease-causing alpha-synuclein variant A30P bound less efficiently to anionic phospholipids, while the variant E46K showed enhanced binding. This suggests that the natural association of alpha-synuclein with membranes is altered in the inherited forms of Parkinson's disease.
Collapse
Affiliation(s)
- Martin Stöckl
- Humboldt-Universität zu Berlin, Mathematisch-Naturwissenschaftliche Fakultät I, Institut für Biologie/Biophysik, Invalidenstr. 43, D-10115 Berlin, Germany
| | | | | | | |
Collapse
|