1
|
Arshad H, Eid S, Mehra S, Williams D, Kaczmarczyk L, Stuart E, Jackson WS, Schmitt-Ulms G, Watts JC. The brain interactome of a permissive prion replication substrate. Neurobiol Dis 2025; 206:106802. [PMID: 39800229 DOI: 10.1016/j.nbd.2025.106802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/10/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025] Open
Abstract
Bank voles are susceptible to prion strains from many different species, yet the molecular mechanisms underlying the ability of bank vole prion protein (BVPrP) to function as a universal prion acceptor remain unclear. Potential differences in molecular environments and protein interaction networks on the cell surface of brain cells may contribute to BVPrP's unusual behavior. To test this hypothesis, we generated knock-in mice that express physiological levels of BVPrP (M109 isoform) and employed mass spectrometry to compare the interactomes of mouse (Mo) PrP and BVPrP following mild in vivo crosslinking of brain tissue. Substantial overlap was observed between the top interactors for BVPrP and MoPrP, with established PrP-interactors such as neural cell adhesion molecules, subunits of Na+/K+-ATPases, and contactin-1 being equally present in the two interactomes. We conclude that the molecular environments of BVPrP and MoPrP in the brains of mice are very similar. This suggests that the unorthodox properties of BVPrP are unlikely to be mediated by differential interactions with other proteins.
Collapse
Affiliation(s)
- Hamza Arshad
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Shehab Eid
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Surabhi Mehra
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Lech Kaczmarczyk
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Erica Stuart
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Walker S Jackson
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Iwaide S, Murakami T, Sedghi Masoud N, Kobayashi N, Fortin JS, Miyahara H, Higuchi K, Chambers JK. Classification of amyloidosis and protein misfolding disorders in animals 2024: A review on pathology and diagnosis. Vet Pathol 2025; 62:117-138. [PMID: 39389927 DOI: 10.1177/03009858241283750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Amyloidosis is a group of diseases in which proteins become amyloid, an insoluble fibrillar aggregate, resulting in organ dysfunction. Amyloid deposition has been reported in various animal species. To diagnose and understand the pathogenesis of amyloidosis, it is important to identify the amyloid precursor protein involved in each disease. Although 42 amyloid precursor proteins have been reported in humans, little is known about amyloidosis in animals, except for a few well-described amyloid proteins, including amyloid A (AA), amyloid light chain (AL), amyloid β (Aβ), and islet amyloid polypeptide-derived amyloid. Recently, several types of novel amyloidosis have been identified in animals using immunohistochemistry and mass spectrometry-based proteomic analysis. Certain species are predisposed to specific types of amyloidosis, suggesting a genetic background for its pathogenesis. Age-related amyloidosis has also emerged due to the increased longevity of captive animals. In addition, experimental studies have shown that some amyloids may be transmissible. Accurate diagnosis and understanding of animal amyloidosis are necessary for appropriate therapeutic intervention and comparative pathological studies. This review provides an updated classification of animal amyloidosis, including associated protein misfolding disorders of the central nervous system, and the current understanding of their pathogenesis. Pathologic features are presented together with state-of-the-art diagnostic methods that can be applied for routine diagnosis and identification of novel amyloid proteins in animals.
Collapse
Affiliation(s)
- Susumu Iwaide
- Tokyo University of Agriculture and Technology, Fuchu-shi, Japan
| | - Tomoaki Murakami
- Tokyo University of Agriculture and Technology, Fuchu-shi, Japan
| | | | | | | | | | - Keiichi Higuchi
- Shinshu University, Matsumoto, Japan
- Meio University, Nago, Japan
| | | |
Collapse
|
3
|
Castle AR, Westaway D. Prion Protein Endoproteolysis: Cleavage Sites, Mechanisms and Connections to Prion Disease. J Neurochem 2025; 169:e16310. [PMID: 39874431 PMCID: PMC11774512 DOI: 10.1111/jnc.16310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
Highly abundant in neurons, the cellular prion protein (PrPC) is an obligatory precursor to the disease-associated misfolded isoform denoted PrPSc that accumulates in the rare neurodegenerative disorders referred to either as transmissible spongiform encephalopathies (TSEs) or as prion diseases. The ability of PrPC to serve as a substrate for this template-mediated conversion process depends on several criteria but importantly includes the presence or absence of certain endoproteolytic events performed at the cell surface or in acidic endolysosomal compartments. The major endoproteolytic events affecting PrPC are referred to as α- and β-cleavages, and in this review we outline the sites within PrPC at which the cleavages occur, the mechanisms potentially responsible and their relevance to pathology. Although the association of α-cleavage with neuroprotection is well-supported, we identify open questions regarding the importance of β-cleavage in TSEs and suggest experimental approaches that could provide clarification. We also combine findings from in vitro cleavage assays and mass spectrometry-based studies of prion protein fragments in the brain to present an updated view in which α- and β-cleavages may represent two distinct clusters of proteolytic events that occur at multiple neighbouring sites rather than at single positions. Furthermore, we highlight the candidate proteolytic mechanisms best supported by the literature; currently, despite several proteases identified as capable of processing PrPC in vitro, in cell-based models and in some cases, in vivo, none have been shown conclusively to cleave PrPC in the brain. Addressing this knowledge gap will be crucial for developing therapeutic interventions to drive PrPC endoproteolysis in a neuroprotective direction. Finally, we end this review by briefly addressing other cleavage events, specifically ectodomain shedding, γ-cleavage, the generation of atypical pathological fragments in the familial prion disorder Gerstmann-Sträussler-Scheinker syndrome and the possibility of an additional form of endoproteolysis close to the PrPC N-terminus.
Collapse
Affiliation(s)
- Andrew R. Castle
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Kavli Institute of Nanoscience DiscoveryUniversity of OxfordOxfordUK
| | - David Westaway
- Centre for Prions and Protein Folding DiseasesUniversity of AlbertaEdmontonCanada
| |
Collapse
|
4
|
Nguyen TTD, Zayed M, Kim YC, Jeong BH. The First Genetic Characterization of the SPRN Gene in Pekin Ducks ( Anas platyrhynchos domesticus). Animals (Basel) 2024; 14:1588. [PMID: 38891635 PMCID: PMC11171214 DOI: 10.3390/ani14111588] [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: 04/27/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Prion diseases are fatal neurodegenerative disorders characterized by an accumulation of misfolded prion protein (PrPSc) in brain tissues. The shadow of prion protein (Sho) encoded by the shadow of prion protein gene (SPRN) is involved in prion disease progress. The interaction between Sho and PrP accelerates the PrPSc conversion rate while the SPRN gene polymorphisms have been associated with prion disease susceptibility in several species. Until now, the SPRN gene has not been investigated in ducks. We identified the duck SPRN gene sequence and investigated the genetic polymorphisms of 184 Pekin ducks. We compared the duck SPRN nucleotide sequence and the duck Sho protein amino acid sequence with those of several other species. Finally, we predicted the duck Sho protein structure and the effects of non-synonymous single nucleotide polymorphisms (SNPs) using computational programs. We were the first to report the Pekin duck SPRN gene sequence. The duck Sho protein sequence showed 100% identity compared with the chicken Sho protein sequence. We found 27 novel SNPs in the duck SPRN gene. Four amino acid substitutions were predicted to affect the hydrogen bond distribution in the duck Sho protein structure. Although MutPred2 and SNPs&GO predicted that all non-synonymous polymorphisms were neutral or benign, SIFT predicted that four variants, A22T, G49D, A68T, and M105I, were deleterious. To the best of our knowledge, this is the first report about the genetic and structural characteristics of the duck SPRN gene.
Collapse
Affiliation(s)
- Thi-Thuy-Duong Nguyen
- Korea Zoonosis Research Institute, Jeonbuk National University, 820-120, Hana-ro, Iksan 54531, Republic of Korea (M.Z.)
- Department of Bioactive Material Sciences, Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Mohammed Zayed
- Korea Zoonosis Research Institute, Jeonbuk National University, 820-120, Hana-ro, Iksan 54531, Republic of Korea (M.Z.)
- Department of Bioactive Material Sciences, Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Department of Surgery, College of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - Yong-Chan Kim
- Department of Biological Sciences, Andong National University, Andong 36729, Republic of Korea
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Jeonbuk National University, 820-120, Hana-ro, Iksan 54531, Republic of Korea (M.Z.)
- Department of Bioactive Material Sciences, Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|
5
|
Castle AR, Kang SG, Eskandari-Sedighi G, Wohlgemuth S, Nguyen MA, Drucker DJ, Mulvihill EE, Westaway D. Beta-endoproteolysis of the cellular prion protein by dipeptidyl peptidase-4 and fibroblast activation protein. Proc Natl Acad Sci U S A 2023; 120:e2209815120. [PMID: 36574660 PMCID: PMC9910601 DOI: 10.1073/pnas.2209815120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/18/2022] [Indexed: 12/29/2022] Open
Abstract
The cellular prion protein (PrPC) converts to alternatively folded pathogenic conformations (PrPSc) in prion infections and binds neurotoxic oligomers formed by amyloid-β α-synuclein, and tau. β-Endoproteolysis, which splits PrPC into N- and C-terminal fragments (N2 and C2, respectively), is of interest because a protease-resistant, C2-sized fragment (C2Sc) accumulates in the brain during prion infections, seemingly comprising the majority of PrPSc at disease endpoint in mice. However, candidates for the underlying proteolytic mechanism(s) remain unconfirmed in vivo. Here, a cell-based screen of protease inhibitors unexpectedly linked type II membrane proteins of the S9B serine peptidase subfamily to PrPC β-cleavage. Overexpression experiments in cells and assays with recombinant proteins confirmed that fibroblast activation protein (FAP) and its paralog, dipeptidyl peptidase-4 (DPP4), cleave directly at multiple sites within PrPC's N-terminal domain. For wild-type mouse and human PrPC substrates expressed in cells, the rank orders of activity were human FAP ~ mouse FAP > mouse DPP4 > human DPP4 and human FAP > mouse FAP > mouse DPP4 >> human DPP4, respectively. C2 levels relative to total PrPC were reduced in several tissues from FAP-null mice, and, while knockout of DPP4 lacked an analogous effect, the combined DPP4/FAP inhibitor linagliptin, but not the FAP-specific inhibitor SP-13786, reduced C2Sc and total PrPSc levels in two murine cell-based models of prion infections. Thus, the net activity of the S9B peptidases FAP and DPP4 and their cognate inhibitors/modulators affect the physiology and pathogenic potential of PrPC.
Collapse
Affiliation(s)
- Andrew R. Castle
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, ABT6G 2M8, Canada
- Department of Medicine, University of Alberta, Edmonton, ABT6G 2G3, Canada
| | - Sang-Gyun Kang
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, ABT6G 2M8, Canada
- Department of Medicine, University of Alberta, Edmonton, ABT6G 2G3, Canada
| | - Ghazaleh Eskandari-Sedighi
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, ABT6G 2M8, Canada
- Department of Biochemistry, University of Alberta, Edmonton, ABT6G 2H7, Canada
| | - Serene Wohlgemuth
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, ABT6G 2M8, Canada
- Department of Medicine, University of Alberta, Edmonton, ABT6G 2G3, Canada
| | - My-Anh Nguyen
- University of Ottawa Heart Institute, Ottawa, ONK1Y 4W7, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ONK1H 8M5, Canada
| | - Daniel J. Drucker
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ONM5G 1X5, Canada
- Department of Medicine, University of Toronto, Toronto, ONM5S 2J7, Canada
| | - Erin E. Mulvihill
- University of Ottawa Heart Institute, Ottawa, ONK1Y 4W7, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ONK1H 8M5, Canada
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, ABT6G 2M8, Canada
- Department of Medicine, University of Alberta, Edmonton, ABT6G 2G3, Canada
- Department of Biochemistry, University of Alberta, Edmonton, ABT6G 2H7, Canada
| |
Collapse
|
6
|
Membrane Domain Localization and Interaction of the Prion-Family Proteins, Prion and Shadoo with Calnexin. MEMBRANES 2021; 11:membranes11120978. [PMID: 34940479 PMCID: PMC8704586 DOI: 10.3390/membranes11120978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 11/30/2022]
Abstract
The cellular prion protein (PrPC) is renowned for its infectious conformational isoform PrPSc, capable of templating subsequent conversions of healthy PrPCs and thus triggering the group of incurable diseases known as transmissible spongiform encephalopathies. Besides this mechanism not being fully uncovered, the protein’s physiological role is also elusive. PrPC and its newest, less understood paralog Shadoo are glycosylphosphatidylinositol-anchored proteins highly expressed in the central nervous system. While they share some attributes and neuroprotective actions, opposing roles have also been reported for the two; however, the amount of data about their exact functions is lacking. Protein–protein interactions and membrane microdomain localizations are key determinants of protein function. Accurate identification of these functions for a membrane protein, however, can become biased due to interactions occurring during sample processing. To avoid such artifacts, we apply a non-detergent-based membrane-fractionation approach to study the prion protein and Shadoo. We show that the two proteins occupy similarly raft and non-raft membrane fractions when expressed in N2a cells and that both proteins pull down the chaperone calnexin in both rafts and non-rafts. These indicate their possible binding to calnexin in both types of membrane domains, which might be a necessary requisite to aid the inherently unstable native conformation during their lifetime.
Collapse
|
7
|
Eskandari-Sedighi G, Cortez LM, Yang J, Daude N, Shmeit K, Sim V, Westaway D. Quaternary Structure Changes for PrP Sc Predate PrP C Downregulation and Neuronal Death During Progression of Experimental Scrapie Disease. Mol Neurobiol 2021; 58:375-390. [PMID: 32959170 PMCID: PMC7695655 DOI: 10.1007/s12035-020-02112-z] [Citation(s) in RCA: 9] [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: 03/28/2020] [Accepted: 09/01/2020] [Indexed: 01/20/2023]
Abstract
Prion diseases are fatal neurodegenerative diseases in mammals with the unique characteristics of misfolding and aggregation of the cellular prion protein (PrPC) to the scrapie prion (PrPSc). Although neuroinflammation and neuronal loss feature within the disease process, the details of PrPC/PrPSc molecular transition to generate different aggregated species, and the correlation between each species and sequence of cellular events in disease pathogenesis are not fully understood. In this study, using mice inoculated with the RML isolate of mouse-adapted scrapie as a model, we applied asymmetric flow field-flow fractionation to monitor PrPC and PrPSc particle sizes and we also measured seeding activity and resistance to proteases. For cellular analysis in brain tissue, we measured inflammatory markers and synaptic damage, and used the isotropic fractionator to measure neuronal loss; these techniques were applied at different timepoints in a cross-sectional study of disease progression. Our analyses align with previous reports defining significant decreases in PrPC levels at pre-clinical stages of the disease and demonstrate that these decreases become significant before neuronal loss. We also identified the earliest PrPSc assemblies at a timepoint equivalent to 40% elapsed time for the disease incubation period; we propose that these assemblies, mostly composed of proteinase K (PK)-sensitive species, play an important role in triggering disease pathogenesis. Lastly, we show that the PK-resistant assemblies of PrPSc that appear at timepoints close to the terminal stage have similar biophysical characteristics, and hence that preparative use of PK-digestion selects for this specific subpopulation. In sum, our data argue that qualitative, as well as quantitative, changes in PrP conformers occur at the midpoint of subclinical phase; these changes affect quaternary structure and may occur at the threshold where adaptive responses become inadequate to deal with pathogenic processes.
Collapse
Affiliation(s)
- Ghazaleh Eskandari-Sedighi
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada
| | - Leonardo M Cortez
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Jing Yang
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada
| | - Nathalie Daude
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada
| | - Klinton Shmeit
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada
| | - Valerie Sim
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - David Westaway
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.
| |
Collapse
|
8
|
Arshad H, Bourkas MEC, Watts JC. The utility of bank voles for studying prion disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:179-211. [PMID: 32958232 DOI: 10.1016/bs.pmbts.2020.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The transmission of prions between species is typically an inefficient process due to the species barrier, which represents incompatibility between prion seed and substrate molecules. Bank voles (Myodes glareolus) are an exception to this rule, as they are susceptible to a diverse range of prion strains from many different animal species. In particular, bank voles can be efficiently infected with most types of human prions and have played a critical role in validating variably protease-sensitive prionopathy (VPSPr) and certain forms of Gerstmann-Sträussler-Scheinker (GSS) disease as bona fide prion disorders rather than non-transmissible proteinopathies. The bank vole prion protein (BVPrP) confers a "universal prion acceptor" phenotype when expressed in mice and when used as a substrate for in vitro prion amplification assays, indicating that the unique prion transmission properties of bank voles are mediated by BVPrP. Over-expression of BVPrP in mice can also promote the spontaneous development of prion disease, indicating that BVPrP is intrinsically prone to both spontaneous and template-directed misfolding. Here, we discuss the utility of bank voles and BVPrP for prion research and how they have provided new tools for establishing rapid animal bioassays, modeling spontaneous prion disease, standardizing prion diagnostics, and understanding the molecular basis of the species barrier.
Collapse
Affiliation(s)
- Hamza Arshad
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Matthew E C Bourkas
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Passet B, Castille J, Makhzami S, Truchet S, Vaiman A, Floriot S, Moazami-Goudarzi K, Vilotte M, Gaillard AL, Helary L, Bertaud M, Andréoletti O, Vaiman D, Calvel P, Daniel-Carlier N, Moudjou M, Beauvallet C, Benharouga M, Laloé D, Mouillet-Richard S, Duchesne A, Béringue V, Vilotte JL. The Prion-like protein Shadoo is involved in mouse embryonic and mammary development and differentiation. Sci Rep 2020; 10:6765. [PMID: 32317725 PMCID: PMC7174383 DOI: 10.1038/s41598-020-63805-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 04/03/2020] [Indexed: 11/09/2022] Open
Abstract
Shadoo belongs to the prion protein family, an evolutionary conserved and extensively studied family due to the implication of PrP in Transmissible Spongiform Encephalopathies. However, the biological function of these genes remains poorly understood. While Sprn-knockdown experiments suggested an involvement of Shadoo during mouse embryonic development, Sprn-knockout experiments in 129Pas/C57BL/6J or 129Pas/FVB/NCr mice did not confirm it. In the present study, we analyzed the impact of Sprn gene invalidation in a pure FVB/NJ genetic background, using a zinc finger nuclease approach. The in-depth analysis of the derived knockout transgenic mice revealed a significant increase in embryonic lethality at early post-implantation stages, a growth retardation of young Sprn-knockout pups fed by wild type mice and a lactation defect of Sprn-knockout females. Histological and transcriptional analyses of knockout E7.5 embryos, E14.5 placentas and G7.5 mammary glands revealed specific roles of the Shadoo protein in mouse early embryogenesis, tissue development and differentiation with a potential antagonist action between PrP and Shadoo. This study thus highlights the entanglement between the proteins of the prion family.
Collapse
Affiliation(s)
- Bruno Passet
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Johan Castille
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Samira Makhzami
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Sandrine Truchet
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Anne Vaiman
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Sandrine Floriot
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | | | - Marthe Vilotte
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Anne-Laure Gaillard
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Louise Helary
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Maud Bertaud
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | | | - Daniel Vaiman
- Institut Cochin, U1016, INSERM, UMR 8504 CNRS, Université de Paris, Paris, France
| | - Pierre Calvel
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | | | - Mohammed Moudjou
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Christian Beauvallet
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | | | - Denis Laloé
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Sophie Mouillet-Richard
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006, Paris, France
| | - Amandine Duchesne
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France
| | - Vincent Béringue
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- Université Paris-Saclay, INRAE, AgroParisTech, UMR1313-GABI, 78350, Jouy-en-Josas, France.
| |
Collapse
|
10
|
Murakami T, Noguchi K, Hachiya N, Kametani F, Tasaki M, Nakaba S, Sassa Y, Yamashita T, Obayashi K, Ando Y, Hamamura M, Kanno T, Kawasako K. Needle-shaped amyloid deposition in rat mammary gland: evidence of a novel amyloid fibril protein. Amyloid 2020; 27:25-35. [PMID: 31615282 DOI: 10.1080/13506129.2019.1675623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Amyloidosis is an extremely rare event in rats. In this study, we report that lipopolysaccharide binding protein (LBP) is the most likely amyloidogenic protein in rat mammary amyloidosis. Histologically, corpora amylacea (CA) and stromal amyloid (SA) were observed in rat mammary glands, and needle-shaped amyloid (NA) was also observed on the surface or gap of CA and SA. Following surveillance in aged rats, NA was observed in 62% of mammary tumours, 25% of male mammary glands and 83% of female mammary glands. Proteomic analysis showed that lactadherin was a major constitutive protein of CA and SA, and both were positive following immunohistochemistry with anti-lactadherin antibodies. In the same analysis, LBP was detected as a prime candidate protein in NA, and NA was positive following immunohistochemistry and immunoelectron microscopy with anti-LBP antibody. Furthermore, synthetic peptides derived from rat LBP formed amyloid fibrils in vitro. Overall, these results provide evidence that LBP is an amyloid precursor protein of NA in rat mammary glands.
Collapse
Affiliation(s)
- Tomoaki Murakami
- Division of Animal Life Science, Graduate School, Institute of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Keiichi Noguchi
- Research Center for Science and Technology, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Naomi Hachiya
- Tokyo Metropolitan Industrial Technology Research Institute, Tokyo, Japan
| | - Fuyuki Kametani
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masayoshi Tasaki
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Morphological and Physiological Sciences, Graduate School of Health Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Nakaba
- Division of Natural Resources and Eco-Materials, Graduate School, Institute of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Yukiko Sassa
- Division of Animal Life Science, Graduate School, Institute of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Taro Yamashita
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Konen Obayashi
- Department of Morphological and Physiological Sciences, Graduate School of Health Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukio Ando
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masao Hamamura
- Pathology Department, Nonclinical Research Center, Drug Development Service Segment, LSI Medicine Corporation, Uto, Japan
| | - Takeshi Kanno
- Pathology Department, Nonclinical Research Center, Drug Development Service Segment, LSI Medicine Corporation, Uto, Japan
| | - Kazufumi Kawasako
- Pathology Department, Nonclinical Research Center, Drug Development Service Segment, LSI Medicine Corporation, Uto, Japan
| |
Collapse
|
11
|
Minikel EV, Kuhn E, Cocco AR, Vallabh SM, Hartigan CR, Reidenbach AG, Safar JG, Raymond GJ, McCarthy MD, O'Keefe R, Llorens F, Zerr I, Capellari S, Parchi P, Schreiber SL, Carr SA. Domain-specific Quantification of Prion Protein in Cerebrospinal Fluid by Targeted Mass Spectrometry. Mol Cell Proteomics 2019; 18:2388-2400. [PMID: 31558565 PMCID: PMC6885701 DOI: 10.1074/mcp.ra119.001702] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/16/2019] [Indexed: 01/11/2023] Open
Abstract
Therapies currently in preclinical development for prion disease seek to lower prion protein (PrP) expression in the brain. Trials of such therapies are likely to rely on quantification of PrP in cerebrospinal fluid (CSF) as a pharmacodynamic biomarker and possibly as a trial endpoint. Studies using PrP ELISA kits have shown that CSF PrP is lowered in the symptomatic phase of disease, a potential confounder for reading out the effect of PrP-lowering drugs in symptomatic patients. Because misfolding or proteolytic cleavage could potentially render PrP invisible to ELISA even if its concentration were constant or increasing in disease, we sought to establish an orthogonal method for CSF PrP quantification. We developed a multi-species targeted mass spectrometry method based on multiple reaction monitoring (MRM) of nine PrP tryptic peptides quantified relative to an isotopically labeled recombinant protein standard for human samples, or isotopically labeled synthetic peptides for nonhuman species. Analytical validation experiments showed process replicate coefficients of variation below 15%, good dilution linearity and recovery, and suitable performance for both CSF and brain homogenate and across humans as well as preclinical species of interest. In n = 55 CSF samples from individuals referred to prion surveillance centers with rapidly progressive dementia, all six human PrP peptides, spanning the N- and C-terminal domains of PrP, were uniformly reduced in prion disease cases compared with individuals with nonprion diagnoses. Thus, lowered CSF PrP concentration in prion disease is a genuine result of the disease process and not an artifact of ELISA-based measurement. As a result, dose-finding studies for PrP lowering drugs may need to be conducted in presymptomatic at-risk individuals rather than in symptomatic patients. We provide a targeted mass spectrometry-based method suitable for preclinical quantification of CSF PrP as a tool for drug development.
Collapse
Affiliation(s)
- Eric Vallabh Minikel
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115; Prion Alliance, Cambridge, MA 02139; Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142.
| | - Eric Kuhn
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115
| | - Alexandra R Cocco
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Sonia M Vallabh
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115; Prion Alliance, Cambridge, MA 02139
| | | | - Andrew G Reidenbach
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Jiri G Safar
- Departments of Pathology and Neurology Case Western Reserve University, Cleveland, OH 44106
| | - Gregory J Raymond
- Laboratory of Persistent Viral Diseases, NIAID Rocky Mountain Labs, Hamilton, MT 59840
| | - Michael D McCarthy
- Environmental Health and Safety, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Rhonda O'Keefe
- Environmental Health and Safety, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Franc Llorens
- National Reference Center for TSE, Georg-August University, Göttingen, 37073, Germany; Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), L'Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Inga Zerr
- National Reference Center for TSE, Georg-August University, Göttingen, 37073, Germany
| | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, 40139, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, 40123, Italy
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, 40139, Italy; Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, 40138, Italy
| | - Stuart L Schreiber
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142; Department of Chemistry & Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Steven A Carr
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142.
| |
Collapse
|
12
|
Proteasomal Inhibition Redirects the PrP-Like Shadoo Protein to the Nucleus. Mol Neurobiol 2019; 56:7888-7904. [PMID: 31129810 PMCID: PMC6815274 DOI: 10.1007/s12035-019-1623-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/24/2019] [Indexed: 01/08/2023]
Abstract
The Shadoo protein (Sho) exhibits homology to the hydrophobic region of the cellular isoform of prion protein (PrPC). As prion-infected brains gradually accumulate infectivity-associated isoforms of prion protein (PrPSc), levels of mature endogenous Sho become reduced. To study the regulatory effect of the proteostatic network on Sho expression, we investigated the action of lactacystin, MG132, NH4Cl, and 3-methyladenine (3-MA) in two cell culture models. In primary mixed neuronal and glial cell cultures (MNGCs) from transgenic mice expressing wild-type Sho from the PrP gene promoter (Tg.Sprn mice), lactacystin- and MG132-mediated inhibition of proteasomal activity shifted the repertoire of Sho species towards unglycosylated forms appearing in the nuclei; conversely, the autophagic modulators NH4Cl and 3-MA did not affect Sho or PrPC glycosylation patterns. Mouse N2a neuroblastoma cells expressing Sho under control of a housekeeping gene promoter treated with MG132 or lactacystin also showed increased nuclear localization of unglycosylated Sho. As two proteasomal inhibitors tested in two cell paradigms caused redirection of Sho to nuclei at the expense of processing through the secretory pathway, our findings define a balanced shift in subcellular localization that thereby differs from the decreases in net Sho species seen in prion-infected brains. Our data are indicative of a physiological pathway to access Sho functions in the nucleus under conditions of impaired proteasomal activity. We also infer that these conditions would comprise a context wherein Sho’s N-terminal nucleic acid–binding RGG repeat region is brought into play.
Collapse
|
13
|
The function of the cellular prion protein in health and disease. Acta Neuropathol 2018; 135:159-178. [PMID: 29151170 DOI: 10.1007/s00401-017-1790-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/11/2022]
Abstract
The essential role of the cellular prion protein (PrPC) in prion disorders such as Creutzfeldt-Jakob disease is well documented. Moreover, evidence is accumulating that PrPC may act as a receptor for protein aggregates and transduce neurotoxic signals in more common neurodegenerative disorders, such as Alzheimer's disease. Although the pathological roles of PrPC have been thoroughly characterized, a general consensus on its physiological function within the brain has not yet been established. Knockout studies in various organisms, ranging from zebrafish to mice, have implicated PrPC in a diverse range of nervous system-related activities that include a key role in the maintenance of peripheral nerve myelination as well as a general ability to protect against neurotoxic stimuli. Thus, the function of PrPC may be multifaceted, with different cell types taking advantage of unique aspects of its biology. Deciphering the cellular function(s) of PrPC and the consequences of its absence is not simply an academic curiosity, since lowering PrPC levels in the brain is predicted to be a powerful therapeutic strategy for the treatment of prion disease. In this review, we outline the various approaches that have been employed in an effort to uncover the physiological and pathological functions of PrPC. While these studies have revealed important clues about the biology of the prion protein, the precise reason for PrPC's existence remains enigmatic.
Collapse
|
14
|
Ricci A, Allende A, Bolton D, Chemaly M, Davies R, Fernández Escámez PS, Gironés R, Herman L, Koutsoumanis K, Lindqvist R, Nørrung B, Robertson L, Ru G, Sanaa M, Skandamis P, Speybroeck N, Simmons M, Kuile BT, Threlfall J, Wahlström H, Acutis PL, Andreoletti O, Goldmann W, Langeveld J, Windig JJ, Ortiz Pelaez A, Snary E. Genetic resistance to transmissible spongiform encephalopathies (TSE) in goats. EFSA J 2017; 15:e04962. [PMID: 32625625 PMCID: PMC7010077 DOI: 10.2903/j.efsa.2017.4962] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Breeding programmes to promote resistance to classical scrapie, similar to those for sheep in existing transmissible spongiform encephalopathies (TSE) regulations, have not been established in goats. The European Commission requested a scientific opinion from EFSA on the current knowledge of genetic resistance to TSE in goats. An evaluation tool, which considers both the weight of evidence and strength of resistance to classical scrapie of alleles in the goat PRNP gene, was developed and applied to nine selected alleles of interest. Using the tool, the quality and certainty of the field and experimental data are considered robust enough to conclude that the K222, D146 and S146 alleles both confer genetic resistance against classical scrapie strains known to occur naturally in the EU goat population, with which they have been challenged both experimentally and under field conditions. The weight of evidence for K222 is greater than that currently available for the D146 and S146 alleles and for the ARR allele in sheep in 2001. Breeding for resistance can be an effective tool for controlling classical scrapie in goats and it could be an option available to member states, both at herd and population levels. There is insufficient evidence to assess the impact of K222, D146 and S146 alleles on susceptibility to atypical scrapie and bovine spongiform encephalopathy (BSE), or on health and production traits. These alleles are heterogeneously distributed across the EU Member States and goat breeds, but often at low frequencies (< 10%). Given these low frequencies, high selection pressure may have an adverse effect on genetic diversity so any breeding for resistance programmes should be developed at Member States, rather than EU level and their impact monitored, with particular attention to the potential for any negative impact in rare or small population breeds.
Collapse
|
15
|
Abstract
Shadoo (Sho), a member of prion protein family, has been shown to prevent embryonic lethality in Prnp0/0 mice and to be reduced in the brains of rodents with terminal prion diseases. Sho can also affect PrP structural dynamics and can increase the prion conversion into its misfolded isoform (PrPSc), which is amyloidogenic and strictly related to expression, intracellular localization and association of PrPC to lipid rafts. We reasoned that if Sho possesses a natural tendency to convert to amyloid-like forms in vitro, it should be able to exhibit “prion-like” properties, such as PK-resistance and aggregation state, also in live cells. We tested this hypothesis, by different approaches in neuronal cells, finding that Sho shows folding properties partially dependent on lipid rafts integrity whose alteration, as well as proteasomal block, regulated generation of intermediate Sho isoforms and exacerbated its misfolding. Moreover, a 18 kDa isoform of Sho, likely bearing the signal peptide, was targeted to mitochondria by interacting with the molecular chaperone TRAP1 which, in turn controlled Sho dual targeting to ER or mitochondria. Our studies contribute to understand the role of molecular chaperones and of PrP-related folding intermediates in “prion-like” conversion.
Collapse
|
16
|
Jayawardena N, Kaur M, Nair S, Malmstrom J, Goldstone D, Negron L, Gerrard JA, Domigan LJ. Amyloid Fibrils from Hemoglobin. Biomolecules 2017; 7:E37. [PMID: 28398221 PMCID: PMC5485726 DOI: 10.3390/biom7020037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/16/2017] [Accepted: 04/05/2017] [Indexed: 12/26/2022] Open
Abstract
Amyloid fibrils are a class of insoluble protein nanofibers that are formed via the self-assembly of a wide range of peptides and proteins. They are increasingly exploited for a broad range of applications in bionanotechnology, such as biosensing and drug delivery, as nanowires, hydrogels, and thin films. Amyloid fibrils have been prepared from many proteins, but there has been no definitive characterization of amyloid fibrils from hemoglobin to date. Here, nanofiber formation was carried out under denaturing conditions using solutions of apo-hemoglobin extracted from bovine waste blood. A characteristic amyloid fibril morphology was confirmed by transmission electron microscopy (TEM) and atomic force microscopy (AFM), with mean fibril dimensions of approximately 5 nm diameter and up to several microns in length. The thioflavin T assay confirmed the presence of β-sheet structures in apo-hemoglobin fibrils, and X-ray fiber diffraction showed the characteristic amyloid cross-β quaternary structure. Apo-hemoglobin nanofibers demonstrated high stability over a range of temperatures (-20 to 80 °C) and pHs (2-10), and were stable in the presence of organic solvents and trypsin, confirming their potential as nanomaterials with versatile applications. This study conclusively demonstrates the formation of amyloid fibrils from hemoglobin for the first time, and also introduces a cost-effective method for amyloid fibril manufacture using meat industry by-products.
Collapse
Affiliation(s)
- Nadishka Jayawardena
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand.
| | - Manmeet Kaur
- School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand.
| | - Smitha Nair
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand.
| | - Jenny Malmstrom
- Department of Chemical and Materials Engineering, The University of Auckland, Auckland 1010, New Zealand.
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand.
| | - David Goldstone
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand.
| | | | - Juliet A Gerrard
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand.
- Department of Chemical and Materials Engineering, The University of Auckland, Auckland 1010, New Zealand.
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand.
- Callaghan Innovation, Lower Hutt 5010, New Zealand.
- School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand.
| | - Laura J Domigan
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand.
| |
Collapse
|
17
|
Mays CE, Soto C. The stress of prion disease. Brain Res 2016; 1648:553-560. [PMID: 27060771 DOI: 10.1016/j.brainres.2016.04.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/01/2016] [Accepted: 04/05/2016] [Indexed: 01/31/2023]
Abstract
Prion diseases are fatal neurodegenerative disorders that include scrapie of sheep, bovine spongiform encephalopathy of cattle, chronic wasting disease of cervids, and Creutzfeldt-Jakob disease (CJD) of humans. The etiology for prion diseases can be infectious, sporadic, or hereditary. However, the common denominator for all types is the formation of a transmissible agent composed of a β-sheet-rich, misfolded version of the host-encoded prion protein (PrPC), known as PrPSc. PrPSc self-replicates through a template-assisted process that converts the α-helical conformation of PrPC into the disease-associated isoform. In parallel with PrPSc accumulation, spongiform change is pathologically observed in the central nervous system, where "holes" appear because of massive neuronal death. Here, we review the cellular pathways triggered in response to PrPSc formation and accumulation. Available data suggest that neuronal dysfunction and death may be caused by what originates as a cellular pro-survival response to chronic PrPSc accumulation. We also discuss what is known about the complex cross-talk between the endoplasmic reticulum stress components and the quality control pathways. Better knowledge about these processes may lead to innovative therapeutic strategies based on manipulating the stress response and its consequences for neurodegeneration. This article is part of a Special Issue entitled SI:ER stress.
Collapse
Affiliation(s)
- Charles E Mays
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas Houston Medical School, Houston, TX 77030, USA
| | - Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas Houston Medical School, Houston, TX 77030, USA.
| |
Collapse
|
18
|
Daude N, Gapeshina H, Dong B, Winship I, Westaway D. Neuroprotective properties of the PrP-like Shadoo glycoprotein assessed in the middle cerebral artery occlusion model of ischemia. Prion 2016; 9:376-93. [PMID: 26516793 PMCID: PMC4964864 DOI: 10.1080/19336896.2015.1105432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Biochemical similarities have been noted between the natively unstructured region of the cellular prion protein, PrPC, and a GPI-linked glycoprotein called Shadoo (Sho); these proteins are encoded by the Prnp and Sprn genes, respectively. Both proteins are expressed in the adult central nervous system and they share overlapping partners, including each other, in interactome studies. As prior studies have ascribed neuroprotective properties to the N-terminal region of PrPC, specifically the octarepeat region, we investigated Sho's neuroprotective properties. To this end we assessed Sho-null (Sprn0/0) and hemizygous (Sprn0/+) mice in the middle cerebral artery occlusion (MCAO) model versus wild type mice and also vs. transgene-rescued Sprn0/0-TgSprn mice. Sprn0/0 mice had a tendency to greater fragility in reaching endpoint and deficits in parameters including infarct volume and neurogenesis, with a reciprocal trend noted in transgene-rescued mice; however these effects did not reach significance. Loss of both PrPC and Sho immunostaining occurred in parallel to neuronal loss on the ipsilateral side of MCAO-lesioned animals; while focal elevations in immunostaining in the penumbra region were sometimes evident for PrPC, they were not noted for Sho. Our studies argue against discernible neuroprotective action of Sho in the genetic backgrounds used for this MCAO paradigm. Whether or not the positively charged N-terminal regions in Sho and PrPC fulfil different roles in vivo remains to be determined.
Collapse
Affiliation(s)
- Nathalie Daude
- a Center for Prion and Protein Folding Diseases; University of Alberta ; Edmonton , AB , Canada
| | - Hristina Gapeshina
- a Center for Prion and Protein Folding Diseases; University of Alberta ; Edmonton , AB , Canada
| | - Bin Dong
- b Neurochemical Research Unit; University of Alberta ; Edmonton , AB , Canada
| | - Ian Winship
- b Neurochemical Research Unit; University of Alberta ; Edmonton , AB , Canada
| | - David Westaway
- a Center for Prion and Protein Folding Diseases; University of Alberta ; Edmonton , AB , Canada
| |
Collapse
|
19
|
Zhao H, Wang SQ, Qing LL, Liu LL, Zhang YP. Expression of BSE-associated proteins in the CNS and lymphoreticular tissues of cattle and buffalo. Sci Bull (Beijing) 2016. [DOI: 10.1007/s11434-016-1130-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Nyeste A, Bencsura P, Vida I, Hegyi Z, Homolya L, Fodor E, Welker E. Expression of the Prion Protein Family Member Shadoo Causes Drug Hypersensitivity That Is Diminished by the Coexpression of the Wild Type Prion Protein. J Biol Chem 2016; 291:4473-86. [PMID: 26721882 DOI: 10.1074/jbc.m115.679035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Indexed: 11/06/2022] Open
Abstract
The prion protein (PrP) seems to exert both neuroprotective and neurotoxic activities. The toxic activities are associated with the C-terminal globular parts in the absence of the flexible N terminus, specifically the hydrophobic domain (HD) or the central region (CR). The wild type prion protein (PrP-WT), having an intact flexible part, exhibits neuroprotective qualities by virtue of diminishing many of the cytotoxic effects of these mutant prion proteins (PrPΔHD and PrPΔCR) when coexpressed. The prion protein family member Doppel, which possesses a three-dimensional fold similar to the C-terminal part of PrP, is also harmful to neuronal and other cells in various models, a phenotype that can also be eliminated by the coexpression of PrP-WT. In contrast, another prion protein family member, Shadoo (Sho), a natively disordered protein possessing structural features similar to the flexible N-terminal tail of PrP, exhibits PrP-WT-like protective properties. Here, we report that, contrary to expectations, Sho expression in SH-SY5Y or HEK293 cells induces the same toxic phenotype of drug hypersensitivity as PrPΔCR. This effect is exhibited in a dose-dependent manner and is also counteracted by the coexpression of PrP-WT. The opposing effects of Shadoo in different model systems revealed here may be explored to help discern the relationship of the various toxic activities of mutant PrPs with each other and the neurotoxic effects seen in neurodegenerative diseases, such as transmissible spongiform encephalopathy and Alzheimer disease.
Collapse
Affiliation(s)
- Antal Nyeste
- From the Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary
| | - Petra Bencsura
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| | - István Vida
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and the Institute of Chemistry, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Zoltán Hegyi
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| | - László Homolya
- the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| | - Elfrieda Fodor
- From the Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary
| | - Ervin Welker
- From the Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Hungary, the Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary, and
| |
Collapse
|
21
|
Lassé M, Ulluwishewa D, Healy J, Thompson D, Miller A, Roy N, Chitcholtan K, Gerrard JA. Evaluation of protease resistance and toxicity of amyloid-like food fibrils from whey, soy, kidney bean, and egg white. Food Chem 2016; 192:491-8. [PMID: 26304377 DOI: 10.1016/j.foodchem.2015.07.044] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 07/06/2015] [Accepted: 07/09/2015] [Indexed: 11/12/2022]
Abstract
The structural properties of amyloid fibrils combined with their highly functional surface chemistry make them an attractive new food ingredient, for example as highly effective gelling agents. However, the toxic role of amyloid fibrils in disease may cause some concern about their food safety because it has not been established unequivocally if consumption of food fibrils poses a health risk to consumers. Here we present a study of amyloid-like fibrils from whey, kidney bean, soy bean, and egg white to partially address this concern. Fibrils showed varied resistance to proteolytic digestion in vitro by either Proteinase K, pepsin or pancreatin. The toxicity of mature fibrils was measured in vitro and compared to native protein, early-stage-fibrillar protein, and sonicated fibrils in two immortalised human cancer cell lines, Caco-2 and Hec-1a. There was no reduction in the viability of either Caco-2 or Hec-1a cells after treatment with a fibril concentration of up to 0.25 mg/mL.
Collapse
Affiliation(s)
- Moritz Lassé
- Riddet Institute, Massey University, Palmerston North, New Zealand; Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand; School of Biological Sciences, University of Canterbury, Christchurch, New Zealand.
| | - Dulantha Ulluwishewa
- Riddet Institute, Massey University, Palmerston North, New Zealand; Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
| | - Jackie Healy
- Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand; School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Dion Thompson
- Protein Science & Engineering Team, Callaghan Innovation, Christchurch and Lower Hutt, New Zealand
| | - Antonia Miller
- Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand; Protein Science & Engineering Team, Callaghan Innovation, Christchurch and Lower Hutt, New Zealand
| | - Nicole Roy
- Riddet Institute, Massey University, Palmerston North, New Zealand; Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | - Juliet A Gerrard
- Riddet Institute, Massey University, Palmerston North, New Zealand; Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand; Protein Science & Engineering Team, Callaghan Innovation, Christchurch and Lower Hutt, New Zealand; School of Biological Sciences and School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Li Q, Richard CA, Moudjou M, Vidic J. Purification and Refolding to Amyloid Fibrils of (His)6-tagged Recombinant Shadoo Protein Expressed as Inclusion Bodies in E. coli. J Vis Exp 2015:e53432. [PMID: 26709825 DOI: 10.3791/53432] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The Escherichia coli expression system is a powerful tool for the production of recombinant eukaryotic proteins. We use it to produce Shadoo, a protein belonging to the prion family. A chromatographic method for the purification of (His)6-tagged recombinant Shadoo expressed as inclusion bodies is described. The inclusion bodies are solubilized in 8 M urea and bound to a Ni(2+)-charged column to perform ion affinity chromatography. Bound proteins are eluted by a gradient of imidazole. Fractions containing Shadoo protein are subjected to size exclusion chromatography to obtain a highly purified protein. In the final step purified Shadoo is desalted to remove salts, urea and imidazole. Recombinant Shadoo protein is an important reagent for biophysical and biochemical studies of protein conformation disorders occurring in prion diseases. Many reports demonstrated that prion neurodegenerative diseases originate from the deposition of stable, ordered amyloid fibrils. Sample protocols describing how to fibrillate Shadoo into amyloid fibrils at acidic and neutral/basic pHs are presented. The methods on how to produce and fibrillate Shadoo can facilitate research in laboratories working on prion diseases, since it allows for production of large amounts of protein in a rapid and low cost manner.
Collapse
|
23
|
Prion Infectivity Plateaus and Conversion to Symptomatic Disease Originate from Falling Precursor Levels and Increased Levels of Oligomeric PrPSc Species. J Virol 2015; 89:12418-26. [PMID: 26423957 DOI: 10.1128/jvi.02142-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 09/25/2015] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED In lethal prion neurodegenerative diseases, misfolded prion proteins (PrP(Sc)) replicate by redirecting the folding of the cellular prion glycoprotein (PrP(C)). Infections of different durations can have a subclinical phase with constant levels of infectious particles, but the mechanisms underlying this plateau and a subsequent exit to overt clinical disease are unknown. Using tandem biophysical techniques, we show that attenuated accumulation of infectious particles in presymptomatic disease is preceded by a progressive fall in PrP(C) level, which constricts replication rate and thereby causes the plateau effect. Furthermore, disease symptoms occurred at the threshold associated with increasing levels of small, relatively less protease-resistant oligomeric prion particles (oPrP(Sc)). Although a hypothetical lethal isoform of PrP cannot be excluded, our data argue that diminishing residual PrP(C) levels and continuously increasing levels of oPrP(Sc) are crucial determinants in the transition from presymptomatic to symptomatic prion disease. IMPORTANCE Prions are infectious agents that cause lethal brain diseases; they arise from misfolding of a cell surface protein, PrP(C) to a form called PrP(Sc). Prion infections can have long latencies even though there is no protective immune response. Accumulation of infectious prion particles has been suggested to always reach the same plateau in the brain during latent periods, with clinical disease only occurring when hypothetical toxic forms (called PrP(L) or TPrP) begin to accumulate. We show here that infectivity plateaus arise because PrP(C) precursor levels become downregulated and that the duration of latent periods can be accounted for by the level of residual PrP(C), which transduces a toxic effect, along with the amount of oligomeric forms of PrP(Sc).
Collapse
|
24
|
Qing LL, Zhao H, Liu LL. Progress on low susceptibility mechanisms of transmissible spongiform encephalopathies. DONG WU XUE YAN JIU = ZOOLOGICAL RESEARCH 2015; 35:436-45. [PMID: 25297084 DOI: 10.13918/j.issn.2095-8137.2014.5.436] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs), also known as prion diseases, are a group of fatal neurodegenerative diseases detected in a wide range of mammalian species. The "protein-only" hypothesis of TSE suggests that prions are transmissible particles devoid of nucleic acid and the primary pathogenic event is thought to be the conversion of cellular prion protein (PrP(C)) into the disease-associated isoform (PrP(Sc)). According to susceptibility to TSEs, animals can be classified into susceptible species and low susceptibility species. In this review we focus on several species with low susceptibility to TSEs: dogs, rabbits, horses and buffaloes. We summarize recent studies into the characteristics of low susceptibility regarding protein structure, and biochemical and genetic properties.
Collapse
Affiliation(s)
- Li-Li Qing
- Laboratory of Conservation and Utilization of Bio-resources, Yunnan University, Kunming 650091, China
| | - Hui Zhao
- Laboratory of Conservation and Utilization of Bio-resources, Yunnan University, Kunming 650091, China.
| | - Lin-Lin Liu
- Laboratory of Conservation and Utilization of Bio-resources, Yunnan University, Kunming 650091, China
| |
Collapse
|
25
|
Sakudo A, Onodera T. Prion protein (PrP) gene-knockout cell lines: insight into functions of the PrP. Front Cell Dev Biol 2015; 2:75. [PMID: 25642423 PMCID: PMC4295555 DOI: 10.3389/fcell.2014.00075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/22/2014] [Indexed: 11/13/2022] Open
Abstract
Elucidation of prion protein (PrP) functions is crucial to fully understand prion diseases. A major approach to studying PrP functions is the use of PrP gene-knockout (Prnp (-/-)) mice. So far, six types of Prnp (-/-) mice have been generated, demonstrating the promiscuous functions of PrP. Recently, other PrP family members, such as Doppel and Shadoo, have been found. However, information obtained from comparative studies of structural and functional analyses of these PrP family proteins do not fully reveal PrP functions. Recently, varieties of Prnp (-/-) cell lines established from Prnp (-/-) mice have contributed to the analysis of PrP functions. In this mini-review, we focus on Prnp (-/-) cell lines and summarize currently available Prnp (-/-) cell lines and their characterizations. In addition, we introduce the recent advances in the methodology of cell line generation with knockout or knockdown of the PrP gene. We also discuss how these cell lines have provided valuable insights into PrP functions and show future perspectives.
Collapse
Affiliation(s)
- Akikazu Sakudo
- Laboratory of Biometabolic Chemistry, Faculty of Medicine, School of Health Sciences, University of the Ryukyus Nishihara, Japan
| | - Takashi Onodera
- Research Center for Food Safety, School of Agricultural and Life Sciences, University of Tokyo Tokyo, Japan
| |
Collapse
|
26
|
Li S, Ju C, Han C, Li Z, Liu W, Ye X, Xu J, Xulong L, Wang X, Chen Z, Meng K, Wan J. Unchanged survival rates of Shadoo knockout mice after infection with mouse-adapted scrapie. Prion 2014; 8:339-43. [PMID: 25495671 DOI: 10.4161/19336896.2014.971574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Previous studies have demonstrated that Shadoo (Sho), a GPI-linked glycoprotein encoded by the Sprn gene with a membrane localization similar to PrP(C), is reduced in the brains of rodents with terminal prion disease. To determine the functional significance of Sho in prion disease pathogenesis, Sho-deficient mice were generated by gene targeting. Sho knockout and control wild-type (WT) mice were infected with themouse-adapted scrapie strains 22L or RML. No significant differences in survival, the incubation period of prion disease or other disease features were observed between Sho mutant and WT mice. In this model of prion disease, Sho removal had no effect on disease pathogenesis.
Collapse
Affiliation(s)
- Sha Li
- a Institute of Military Veterinary; Academy of Military Medical Science ; Changchun , China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kaur M, Healy J, Vasudevamurthy M, Lassé M, Puskar L, Tobin MJ, Valery C, Gerrard JA, Sasso L. Stability and cytotoxicity of crystallin amyloid nanofibrils. NANOSCALE 2014; 6:13169-78. [PMID: 25255060 DOI: 10.1039/c4nr04624b] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Previous work has identified crystallin proteins extracted from fish eye lenses as a cheap and readily available source for the self-assembly of amyloid nanofibrils. However, before exploring potential applications, the biophysical aspects and safety of this bionanomaterial need to be assessed so as to ensure that it can be effectively and safely used. In this study, crude crystallin amyloid fibrils are shown to be stable across a wide pH range, in a number of industrially relevant solvents, at both low and high temperatures, and in the presence of proteases. Crystallin nanofibrils were compared to well characterised insulin and whey protein fibrils using Thioflavin T assays and TEM imaging. Cell cytotoxicity assays suggest no adverse impact of both mature and fragmented crystallin fibrils on cell viability of Hec-1a endometrial cells. An IR microspectroscopy study supports long-term structural integrity of crystallin nanofibrils.
Collapse
Affiliation(s)
- Manmeet Kaur
- Biomolecular Interaction Centre and School of Biological Sciences, University of Canterbury, Christchurch 8140, New Zealand.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Watts JC, Giles K, Patel S, Oehler A, DeArmond SJ, Prusiner SB. Evidence that bank vole PrP is a universal acceptor for prions. PLoS Pathog 2014; 10:e1003990. [PMID: 24699458 PMCID: PMC3974871 DOI: 10.1371/journal.ppat.1003990] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 01/28/2014] [Indexed: 12/29/2022] Open
Abstract
Bank voles are uniquely susceptible to a wide range of prion strains isolated from many different species. To determine if this enhanced susceptibility to interspecies prion transmission is encoded within the sequence of the bank vole prion protein (BVPrP), we inoculated Tg(M109) and Tg(I109) mice, which express BVPrP containing either methionine or isoleucine at polymorphic codon 109, with 16 prion isolates from 8 different species: humans, cattle, elk, sheep, guinea pigs, hamsters, mice, and meadow voles. Efficient disease transmission was observed in both Tg(M109) and Tg(I109) mice. For instance, inoculation of the most common human prion strain, sporadic Creutzfeldt-Jakob disease (sCJD) subtype MM1, into Tg(M109) mice gave incubation periods of ∼200 days that were shortened slightly on second passage. Chronic wasting disease prions exhibited an incubation time of ∼250 days, which shortened to ∼150 days upon second passage in Tg(M109) mice. Unexpectedly, bovine spongiform encephalopathy and variant CJD prions caused rapid neurological dysfunction in Tg(M109) mice upon second passage, with incubation periods of 64 and 40 days, respectively. Despite the rapid incubation periods, other strain-specified properties of many prion isolates—including the size of proteinase K–resistant PrPSc, the pattern of cerebral PrPSc deposition, and the conformational stability—were remarkably conserved upon serial passage in Tg(M109) mice. Our results demonstrate that expression of BVPrP is sufficient to engender enhanced susceptibility to a diverse range of prion isolates, suggesting that BVPrP may be a universal acceptor for prions. Prions are infectious proteins that cause devastating neurodegenerative diseases in both humans and animals. Unlike other rodents, bank voles are highly susceptible to prions from many different species, suggesting that bank voles do not impose a “species barrier,” which normally restricts the transmission of prions from one species to another. We were curious as to whether the unprecedented promiscuity of bank voles for prions is due to the specific prion protein sequence expressed, or to some other factor inherent to bank vole physiology. To answer this question, we inoculated transgenic mice that express bank vole prion protein [Tg(BVPrP) mice] with a diverse set of prions deriving from eight different species. Like bank voles, Tg(BVPrP) mice were highly susceptible to prions from all species tested, demonstrating that the BVPrP sequence mediates the enhanced susceptibility of bank voles to prions. Because the amino acid sequences of mouse and BVPrP differ at only eight positions, our results demonstrate that alterations to a small subset of residues within PrP can have a profound effect on the susceptibility of an organism to prions from another species.
Collapse
Affiliation(s)
- Joel C Watts
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California, United States of America; Department of Neurology, University of California, San Francisco, San Francisco, California, United States of America
| | - Kurt Giles
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California, United States of America; Department of Neurology, University of California, San Francisco, San Francisco, California, United States of America
| | - Smita Patel
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California, United States of America
| | - Abby Oehler
- Department of Pathology, University of California, San Francisco, San Francisco, California, United States of America
| | - Stephen J DeArmond
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California, United States of America; Department of Pathology, University of California, San Francisco, San Francisco, California, United States of America
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, California, United States of America; Department of Neurology, University of California, San Francisco, San Francisco, California, United States of America
| |
Collapse
|
29
|
Wang S, Zhao H, Zhang Y. Advances in research on Shadoo, shadow of prion protein. CHINESE SCIENCE BULLETIN-CHINESE 2014. [DOI: 10.1007/s11434-014-0129-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
30
|
Mays CE, Kim C, Haldiman T, van der Merwe J, Lau A, Yang J, Grams J, Di Bari MA, Nonno R, Telling GC, Kong Q, Langeveld J, McKenzie D, Westaway D, Safar JG. Prion disease tempo determined by host-dependent substrate reduction. J Clin Invest 2014; 124:847-58. [PMID: 24430187 DOI: 10.1172/jci72241] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 11/07/2013] [Indexed: 01/01/2023] Open
Abstract
The symptoms of prion infection can take years or decades to manifest following the initial exposure. Molecular markers of prion disease include accumulation of the misfolded prion protein (PrPSc), which is derived from its cellular precursor (PrPC), as well as downregulation of the PrP-like Shadoo (Sho) glycoprotein. Given the overlapping cellular environments for PrPC and Sho, we inferred that PrPC levels might also be altered as part of a host response during prion infection. Using rodent models, we found that, in addition to changes in PrPC glycosylation and proteolytic processing, net reductions in PrPC occur in a wide range of prion diseases, including sheep scrapie, human Creutzfeldt-Jakob disease, and cervid chronic wasting disease. The reduction in PrPC results in decreased prion replication, as measured by the protein misfolding cyclic amplification technique for generating PrPSc in vitro. While PrPC downregulation is not discernible in animals with unusually short incubation periods and high PrPC expression, slowly evolving prion infections exhibit downregulation of the PrPC substrate required for new PrPSc synthesis and as a receptor for pathogenic signaling. Our data reveal PrPC downregulation as a previously unappreciated element of disease pathogenesis that defines the extensive, presymptomatic period for many prion strains.
Collapse
|
31
|
Zhang J, Guo Y, Xie WL, Xu Y, Ren K, Shi Q, Zhang BY, Chen C, Tian C, Gao C, Dong XP. Disruption of glycosylation enhances ubiquitin-mediated proteasomal degradation of Shadoo in Scrapie-infected rodents and cultured cells. Mol Neurobiol 2014; 49:1373-84. [PMID: 24390475 DOI: 10.1007/s12035-013-8612-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 12/08/2013] [Indexed: 01/06/2023]
Abstract
Shadoo (Sho) is an N-glycosylated glycophosphatidylinositol-anchored protein that is expressed in the brain and exhibits neuroprotective properties. Recently, research has shown that a reduction of Sho levels may reflect the presence of PrPSc in the brain. However, the possible mechanism by which prion infection triggers down-regulation of Sho remains unclear. In the present study, Western blot and immunohistochemical assays revealed that Sho, especially glycosylated Sho, declined markedly in the brains of five scrapie agent-infected hamsters and mice at the terminal stages. Analyses of the down-regulation of Sho levels with the emergence of PrPSc C2 proteolytic fragments did not identify close association in all tested scrapie-infected models. To further investigate the mechanism of depletion of Sho in prion disease, a Sho-expressing plasmid with HA tag was introduced into a scrapie-infected cell line, SMB-S15, and its normal cell line, SMB-PS. Western blot assay revealed dramatically decreased Sho in SMB-S15 cells, especially its glycosylated form. Proteasome inhibitor MG132 reversed the decrease of nonglycosylated Sho, but had little effect on glycosylated Sho. N-acetylglucosamine transferase inhibitor tunicamycin efficiently reduced the glycosylations of Sho and PrPC in SMB-PS cells, while two other endoplasmic reticulum stress inducers showed clear inhibition of diglycosylated PrPC, but did not change the expression level and profile of Sho. Furthermore, immunoprecipitation of HA-Sho illustrated ubiquitination of Sho in SMB-S15 cells, but not in SMB-PS cells. We propose that the depletions of Sho in scrapie-infected cell lines due to inhibition of glycosylation mediate protein destabilization and subsequently proteasome degradation after modification by ubiquitination.
Collapse
Affiliation(s)
- Jin Zhang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing, 102206, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mays CE, Coomaraswamy J, Watts JC, Yang J, Ko KW, Strome B, Mercer RC, Wohlgemuth SL, Schmitt-Ulms G, Westaway D. Endoproteolytic processing of the mammalian prion glycoprotein family. FEBS J 2013; 281:862-76. [DOI: 10.1111/febs.12654] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/25/2013] [Accepted: 11/19/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Charles E. Mays
- Centre for Prions and Protein Folding Diseases; University of Alberta; Edmonton Canada
| | | | - Joel C. Watts
- Department of Biochemistry and Tanz Centre for Research in Neurodegenerative Diseases; University of Toronto; ON Canada
| | - Jing Yang
- Centre for Prions and Protein Folding Diseases; University of Alberta; Edmonton Canada
| | - Kerry W.S. Ko
- Centre for Prions and Protein Folding Diseases; University of Alberta; Edmonton Canada
| | - Bob Strome
- Department of Biochemistry and Tanz Centre for Research in Neurodegenerative Diseases; University of Toronto; ON Canada
| | - Robert C.C. Mercer
- Centre for Prions and Protein Folding Diseases; University of Alberta; Edmonton Canada
| | - Serene L. Wohlgemuth
- Centre for Prions and Protein Folding Diseases; University of Alberta; Edmonton Canada
| | - Gerold Schmitt-Ulms
- Department of Biochemistry and Tanz Centre for Research in Neurodegenerative Diseases; University of Toronto; ON Canada
| | - David Westaway
- Centre for Prions and Protein Folding Diseases; University of Alberta; Edmonton Canada
- Division of Neurology; Department of Biochemistry; University of Alberta; Edmonton Canada
| |
Collapse
|
33
|
Li Q, Chevalier C, Henry C, Richard CA, Moudjou M, Vidic J. Shadoo binds lipid membranes and undergoes aggregation and fibrillization. Biochem Biophys Res Commun 2013; 438:519-25. [PMID: 23911790 DOI: 10.1016/j.bbrc.2013.07.104] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 07/25/2013] [Indexed: 10/26/2022]
Abstract
Lipid membrane can enhance prion protein (PrP) pathological fibrillogenesis. A neuronal paralog of PrP, named Shadoo (Sho), is localized to similar membrane environment as PrP and can also convert to amyloid-like fibrilles. To gain insight into the role of Sho in prion diseases, we studied Sho interactions with cellular membrane models. Sho was found to bind anionic lipid vesicles. Spectroscopic and microscopic data showed that membrane-associated Sho slowly converted into amyloid fibers. Furthermore, binding of Sho to anionic liposomes has a disruptive effect on the integrity of the lipid bilayer leading to the formation of supramolecular lipid-protein complexes. In consequence, the role of Sho in prion diseases might depend on the oligomerization state of Sho but also the nature of these lipoprotein assembles.
Collapse
Affiliation(s)
- Qiaojing Li
- Virologie et Immunologie Moléculaires - INRA, Jouy en Josas, France
| | | | | | | | | | | |
Collapse
|
34
|
Introduction: Invited Speakers. Prion 2013; 7:1-9. [PMID: 29095078 PMCID: PMC4031666 DOI: 10.4161/pri.26105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The Alberta Prion Research Institute, part of Alberta Innovates Bio Solutions, is proud to host the world's largest international prion research congress, PRION 2013: Conquering Frontiers, in Banff, Alberta, Canada from May 26-29, 2013. PRION 2013 will be only the second time this international meeting has been held outside of Europe since it began in 2004. The PRION 2013 International Scientific Advisory Committee includes leading international scholars and policy advisors in both human and animal protein misfolding research from 12 countries: Australia, Brazil, Canada, China, England, France, Germany, Japan, The Netherlands, Scotland, Spain and the United States. Prion and protein misfolding science can inform policy, risk management and mitigation, diagnoses and potential treatments in a range of areas from wildlife management to human dementias and neurodegenerative diseases. Compelling evidence is emerging that prion-like mechanisms may underlie a number of the human neurodegenerative diseases and dementias, providing the opportunity to seek out new treatments and for the cross-fertilization of ideas between the two related fields. This approach will be highlighted at PRION 2013. The theme of PRION 2013 is "Conquering Frontiers." It will be a continuation of the science covered in previous meetings with an emphasis on looking toward investigations in the new frontiers created by the relationships between prion diseases and human neurodegenerative diseases and dementias. The four-day session features scientific talks, workshops and posters on the following themes: Prion and Prion-like Diseases in Humans; Prion Diseases in Animals; Protein Structure and Biology; and Socioeconomic Impacts. The knowledge exchange that will take place at PRION 2013 will help to shape the future of prion and protein misfolding research and its application around the world.
Collapse
|
35
|
PrP octarepeats region determined the interaction with caveolin-1 and phosphorylation of caveolin-1 and Fyn. Med Microbiol Immunol 2013; 202:215-27. [PMID: 23283514 DOI: 10.1007/s00430-012-0284-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 12/12/2012] [Indexed: 01/15/2023]
Abstract
Caveolin-1 is one of the major constituents of caveolae. Both Cav-1 and PrP are plasma membrane proteins, which show active capacities for molecular interactions with many other proteins or agents, including themselves. Using yeast two-hybrid system and immunoprecipitation, we reconfirmed the molecular interaction between human Cav-1 and PrP. With co-immunoprecipitation tests, PrP(C)-Cav-1 and PrP(Sc)-Cav-1 complexes were identified in the brain homogenates of normal and scrapie agent 263K-infected hamsters, respectively. Transient expression of wild-type PrP (PrP-PG5) in HEK293 cells did not change the situation of Cav-1 and subsequent signal transduction pathways, while cross-linking of the expressed PrP with specific antibody induced remarkable colocalization of PrP and Cav-1 on the plasma membrane and significant increases of phosphorylated Cav-1 and phosphorylated Fyn. With deleted and inserted PrP mutants within octarepeat region, we observed obvious octarepeat-associated phenomena, including lower binding capacity with Cav-1 in vitro, unable to co-localize with Cav-1 in the cells and to induce up-regulation of p-Cav-1 and p-Fyn when removal of octarepeats in the context of full-length PrP. Moreover, we found that treatment on HEK293 cells with fibrous form of recombinant PrP protein led to up-regulating the levels of p-Cav-1 and p-Fyn. Our data here provide strong evidence that octarepeats of PrP are critical for the interaction between PrP and Cav-1. Significant alterations in the cultured cells, either the distributions of PrP and Cav-1 morphologically or the up-regulations of p-Cav-1 and p-Fyn, induced by antibody-mediated cross-linking or fibrous forms of PrP may suggest a possible internalization process of PrP(Sc).
Collapse
|
36
|
Zhao H, Liu LL, Du SH, Wang SQ, Zhang YP. Comparative analysis of the Shadoo gene between cattle and buffalo reveals significant differences. PLoS One 2012; 7:e46601. [PMID: 23071594 PMCID: PMC3468620 DOI: 10.1371/journal.pone.0046601] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 09/01/2012] [Indexed: 12/03/2022] Open
Abstract
Background While prions play a central role in the pathogenesis of transmissible spongiform encephalopathies, the biology of these proteins and the pathophysiology of these diseases remain largely unknown. Since no case of bovine spongiform encephalopathy (BSE) has ever been reported in buffalo despite their phylogenetic proximity to cattle, genetic differences may be driving the different susceptibilities of these two species to BSE. We thus hypothesized that differences in expression of the most recently identified member of the prion family or Shadoo (SPRN) gene may relate to these species-specific differences. Principal Findings We first analyzed and compared the polymorphisms of the SPRN gene (∼4.4 kb), including the putative promoter, coding and 3′ regions, and further verified the entire ORF and putative promoter. This yielded a total of 117 fixed differences, remarkably: 1) a 12-bp insertion/deletion polymorphism in the hydrophobic domain of the cattle but not buffalo gene, introducing a four amino acid expansion/contraction in a series of 5 tandem Ala/Gly-containing repeats; 2) two fixed missense mutations (102Ser→Gly and 119Thr→Ala), and three missense mutations (92Pro>Thr/Met, 122Thr>Ile and 139Arg>Trp) in the coding region presenting different (P<0.05) genotypic and allelic frequency distributions between cattle and buffalo; and, 3) functional luciferase-reporter experiments for the predicted promoter region, consistent with a significantly higher activity in buffalo than cattle. Supporting these findings, immunoblotting revealed higher relative expression levels of Sho protein in cerebrum from buffalo than from cattle. In addition, for cattle, highest Sho expression was detected in obex, as compared to cerebrum or cerebellum. Significance Our findings support Sho as a non-PrP specific marker for prion infections, with obex as the best tissue source for the detection of Sho in TSE rapid tests. Moreover, these discoveries may prove advantageous for further understanding the biology of prion diseases.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Binding Sites
- Brain/metabolism
- Buffaloes/genetics
- Cattle/genetics
- Encephalopathy, Bovine Spongiform/genetics
- Gene Components
- Gene Expression
- Gene Frequency
- Genes, Reporter
- INDEL Mutation
- Luciferases, Renilla/biosynthesis
- Luciferases, Renilla/genetics
- Mutation, Missense
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Organ Specificity
- Polymorphism, Genetic
- Protein Structure, Tertiary
- Repetitive Sequences, Nucleic Acid
- Sequence Analysis, DNA
- Sequence Homology, Nucleic Acid
Collapse
Affiliation(s)
- Hui Zhao
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, People’s Republic of China
| | - Lin-Lin Liu
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, People’s Republic of China
| | - Shou-Hui Du
- School of Life Science, Yunnan University, Kunming, People’s Republic of China
| | - Si-Qi Wang
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, People’s Republic of China
| | - Ya-Ping Zhang
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, People’s Republic of China
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, People’s Republic of China
- * E-mail:
| |
Collapse
|
37
|
Abstract
Shadoo (Sho) is a brain glycoprotein with similarities to the unstructured region of PrPC. Frameshift alleles of the Sho gene, Sprn, are reported in variant Creutzfeldt-Jakob disease (vCJD) patients while Sprn mRNA knockdown in PrP-null (Prnp0/0) embryos produces lethality, advancing Sho as the hypothetical PrP-like “pi” protein. Also, Sho levels are reduced as misfolded PrP accumulates during prion infections. To penetrate these issues we created Sprn null alleles (Daude et al., Proc. Natl. Acad. Sci USA 2012; 109(23): 9035–40). Results from the challenge of Sprn null and TgSprn transgenic mice with rodent-adapted prions coalesce to define downregulation of Sho as a “tracer” for the formation of misfolded PrP. However, classical BSE and rodent-adapted BSE isolates may behave differently, as they do for other facets of the pathogenic process, and this intriguing variation warrants closer scrutiny. With regards to physiological function, double knockout mice (Sprn0/0/Prnp0/0) mice survived to over 600 d of age. This suggests that Sho is not pi, or, given the accumulating data for many activities for PrPC, that the pi hypothesis invoking a discrete signaling pathway to maintain neuronal viability is no longer tenable.
Collapse
Affiliation(s)
- Nathalie Daude
- Centre for Prions and Protein Folding Diseases, University of Alberta Edmonton, AB, Canada
| | | |
Collapse
|
38
|
Association of an indel polymorphism in the 3′UTR of the caprine SPRN gene with scrapie positivity in the central nervous system. J Gen Virol 2012; 93:1620-1623. [DOI: 10.1099/vir.0.041400-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The aim of this study was to analyse the SPRN genes of goats from several scrapie outbreaks in order to detect polymorphisms and to look for association with scrapie occurrence, by an unmatched case–control study. A region of the caprine SPRN gene encompassing the entire ORF and a fragment of the 3′UTR revealed a total of 11 mutations: 10 single-nucleotide polymorphisms and one indel polymorphism. Only two non-synonymous mutations occurring at very low incidence were identified. A significant association with scrapie positivity in the central nervous system was found for an indel polymorphism (602_606insCTCCC) in the 3′UTR. Bioinformatics analyses suggest that this indel may modulate scrapie susceptibility via a microRNA-mediated post-transcriptional mechanism. This is the first study to demonstrate an association between the SPRN gene and goat scrapie. The identified indel may serve as a genetic target other than PRNP to predict disease risk in future genetics-based scrapie-control approaches in goats.
Collapse
|
39
|
Knockout of the prion protein (PrP)-like Sprn gene does not produce embryonic lethality in combination with PrP(C)-deficiency. Proc Natl Acad Sci U S A 2012; 109:9035-40. [PMID: 22619325 DOI: 10.1073/pnas.1202130109] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Sprn gene encodes Shadoo (Sho), a glycoprotein with biochemical properties similar to the unstructured region of cellular prion protein (PrP(C)). Sho has been considered a candidate for the hypothetical π protein that supplies a PrP(C)-like function to maintain the viability of Prnp(0/0) mice lacking the PrP(C) protein. To understand these relationships more clearly we probed the cell biology of Sho and created knockout mice. Besides full-length and a "C1" C-terminal fragment, we describe a 6-kDa N-terminal Sho neuropeptide, "N1," which is present in membrane-enriched subcellular fractions of wild-type mice. Sprn null alleles were produced that delete all protein coding sequences yet spare the Mtg1 gene transcription unit that overlaps the Sprn 3' UTR; the resulting mice bred to homozygosity were viable and fertile, although Sprn(0/0) mice maintained in two genetic backgrounds weighed less than wild-type mice. Lack of Sho protein did not affect prion incubation time. Contrasting with lethality reported for knockdown of expression in Prnp(0/0) embryos using lentiviruses targeted against the Sprn 3' UTR, we established that double-knockout mice deficient in both Sho and PrP(C) are fertile and viable up to 690 d of age. Our data reduce the impetus for equating Sho with the notional π protein and are not readily reconciled with hypotheses wherein expression of PrP(C) and Sho are both required for completion of embryogenesis. Alternatively, and in accord with some reports for PrP(C), we infer that Sho's activity will prove germane to the maintenance of neuronal viability in postnatal life.
Collapse
|
40
|
Abstract
The evolutionary origins of vertebrate prion genes had remained elusive until recently when multiple lines of evidence converged to the proposition that members of the prion gene family represent an ancient branch of a larger family of ZIP metal ion transporters. (1) A follow-up investigation which explored the mechanism of evolution in more detail led to the surprising conclusion that the emergence of the prion founder gene likely involved the reverse transcription of a spliced transcript of a LIV-1 ZIP predecessor gene. (2) The objective of this perspective is to discuss the possible significance of this reunion of ZIP and prion gene subfamilies for understanding the biology of the prion protein in health and disease. While a recent review article broadly introduced this area of research, (3) the emphasis here is to comment on some of the more pertinent concepts, experimental paradigms, ongoing developments and challenges.
Collapse
Affiliation(s)
- Sepehr Ehsani
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON Canada
| | | | | | | |
Collapse
|
41
|
Spontaneous generation of rapidly transmissible prions in transgenic mice expressing wild-type bank vole prion protein. Proc Natl Acad Sci U S A 2012; 109:3498-503. [PMID: 22331873 DOI: 10.1073/pnas.1121556109] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Currently, there are no animal models of the most common human prion disorder, sporadic Creutzfeldt-Jakob disease (CJD), in which prions are formed spontaneously from wild-type (WT) prion protein (PrP). Interestingly, bank voles (BV) exhibit an unprecedented promiscuity for diverse prion isolates, arguing that bank vole PrP (BVPrP) may be inherently prone to adopting misfolded conformations. Therefore, we constructed transgenic (Tg) mice expressing WT BVPrP. Tg(BVPrP) mice developed spontaneous CNS dysfunction between 108 and 340 d of age and recapitulated the hallmarks of prion disease, including spongiform degeneration, pronounced astrogliosis, and deposition of alternatively folded PrP in the brain. Brain homogenates of ill Tg(BVPrP) mice transmitted disease to Tg(BVPrP) mice in ∼35 d, to Tg mice overexpressing mouse PrP in under 100 d, and to WT mice in ∼185 d. Our studies demonstrate experimentally that WT PrP can spontaneously form infectious prions in vivo. Thus, Tg(BVPrP) mice may be useful for studying the spontaneous formation of prions, and thus may provide insight into the etiology of sporadic CJD.
Collapse
|
42
|
Westaway D, Genovesi S, Daude N, Brown R, Lau A, Lee I, Mays CE, Coomaraswamy J, Canine B, Pitstick R, Herbst A, Yang J, Ko KWS, Schmitt-Ulms G, Dearmond SJ, McKenzie D, Hood L, Carlson GA. Down-regulation of Shadoo in prion infections traces a pre-clinical event inversely related to PrP(Sc) accumulation. PLoS Pathog 2011; 7:e1002391. [PMID: 22114562 PMCID: PMC3219720 DOI: 10.1371/journal.ppat.1002391] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 10/07/2011] [Indexed: 11/18/2022] Open
Abstract
During prion infections of the central nervous system (CNS) the cellular prion protein, PrP(C), is templated to a conformationally distinct form, PrP(Sc). Recent studies have demonstrated that the Sprn gene encodes a GPI-linked glycoprotein Shadoo (Sho), which localizes to a similar membrane environment as PrP(C) and is reduced in the brains of rodents with terminal prion disease. Here, analyses of prion-infected mice revealed that down-regulation of Sho protein was not related to Sprn mRNA abundance at any stage in prion infection. Down-regulation was robust upon propagation of a variety of prion strains in Prnp(a) and Prnp(b) mice, with the exception of the mouse-adapted BSE strain 301 V. In addition, Sho encoded by a TgSprn transgene was down-regulated to the same extent as endogenous Sho. Reduced Sho levels were not seen in a tauopathy, in chemically induced spongiform degeneration or in transgenic mice expressing the extracellular ADan amyloid peptide of familial Danish dementia. Insofar as prion-infected Prnp hemizygous mice exhibited accumulation of PrP(Sc) and down-regulation of Sho hundreds of days prior to onset of neurologic symptoms, Sho depletion can be excluded as an important trigger for clinical disease or as a simple consequence of neuronal damage. These studies instead define a disease-specific effect, and we hypothesize that membrane-associated Sho comprises a bystander substrate for processes degrading PrP(Sc). Thus, while protease-resistant PrP detected by in vitro digestion allows post mortem diagnosis, decreased levels of endogenous Sho may trace an early response to PrP(Sc) accumulation that operates in the CNS in vivo. This cellular response may offer new insights into the homeostatic mechanisms involved in detection and clearance of the misfolded proteins that drive prion disease pathogenesis.
Collapse
Affiliation(s)
- David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|