1
|
RG C, Tallon A, Latch EK. Chronic Wasting Disease Research in North America: A systematic review highlighting species-wise and interdisciplinary research trends. Prion 2025; 19:1-16. [PMID: 39960789 PMCID: PMC11834482 DOI: 10.1080/19336896.2025.2464753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/02/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Chronic Wasting Disease (CWD) research has experienced significant growth, spanning diverse disciplines such as genetics, immunology, modelling, and behaviour. To gain a broad understanding of the changes in CWD research focusing cervids, we analysed temporal trends in study location, species, genus investigated, infection types, and population type since the discovery of CWD in 1980s. Our findings indicate that Colorado, USA, published the highest number of articles, followed by Wisconsin, and publication numbers correlated with reported CWD cases in states/provinces. Odocoileus emerged as the most studied genus. Wild populations are studied more commonly than captive populations. Keyword analysis of transmission types shows the discovery of novel transmission modes in the recent past. We also used a novel approach to categorize studies into five themes: field-based, lab-based, math/analytics/modelling-based, management-based, and human dimensions. Overall, most studies captured had a lab-based component. The interdisciplinary or transdisciplinary nature of major disciplines and evolving trends in keywords, particularly the increased reliance on genetics/genomics, accentuate the beginning of using genomics to under and tackle CWD at a fundamental scale. Encapsulated in our analysis, these dynamic changes offer valuable insights for navigating CWD through scientifically informed proactive management decisions in conjunction with existing surveillance efforts not only for the commonly studied species but also for potentially susceptible species.
Collapse
Affiliation(s)
- Chandika RG
- Department of Biological Sciences, University of Wisconsin, Milwaukee, WI, USA
| | - Anaïs Tallon
- Department of Biological Sciences, University of Wisconsin, Milwaukee, WI, USA
- Marine Conservation Group, Helmholtz Institute for Functional Marine Biodiversity (HIFMB), Oldenburg, Germany
| | - Emily K. Latch
- Department of Biological Sciences, University of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
2
|
Cehlar O, Njemoga S, Horvath M, Cizmazia E, Bednarikova Z, Barrera EE. Structures of Oligomeric States of Tau Protein, Amyloid-β, α-Synuclein and Prion Protein Implicated in Alzheimer's Disease, Parkinson's Disease and Prionopathies. Int J Mol Sci 2024; 25:13049. [PMID: 39684761 DOI: 10.3390/ijms252313049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
In this review, we focus on the biophysical and structural aspects of the oligomeric states of physiologically intrinsically disordered proteins and peptides tau, amyloid-β and α-synuclein and partly disordered prion protein and their isolations from animal models and human brains. These protein states may be the most toxic agents in the pathogenesis of Alzheimer's and Parkinson's disease. It was shown that oligomers are important players in the aggregation cascade of these proteins. The structural information about these structural states has been provided by methods such as solution and solid-state NMR, cryo-EM, crosslinking mass spectrometry, AFM, TEM, etc., as well as from hybrid structural biology approaches combining experiments with computational modelling and simulations. The reliable structural models of these protein states may provide valuable information for future drug design and therapies.
Collapse
Affiliation(s)
- Ondrej Cehlar
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Stefana Njemoga
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Marian Horvath
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Erik Cizmazia
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Zuzana Bednarikova
- Institute of Experimental Physics, Slovak Academy of Sciences, 04001 Kosice, Slovakia
| | - Exequiel E Barrera
- Instituto de Histología y Embriología (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CC56, Universidad Nacional de Cuyo, Mendoza M5502JMA, Argentina
| |
Collapse
|
3
|
Gojanovich AD, Le NTT, Mercer RCC, Park S, Wu B, Anane A, Vultaggio JS, Mostoslavsky G, Harris DA. Abnormal synaptic architecture in iPSC-derived neurons from a multi-generational family with genetic Creutzfeldt-Jakob disease. Stem Cell Reports 2024; 19:1474-1488. [PMID: 39332406 PMCID: PMC11561462 DOI: 10.1016/j.stemcr.2024.08.010] [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: 11/08/2022] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/29/2024] Open
Abstract
Genetic prion diseases are caused by mutations in PRNP, which encodes the prion protein (PrPC). Why these mutations are pathogenic, and how they alter the properties of PrPC are poorly understood. We have consented and accessed 22 individuals of a multi-generational Israeli family harboring the highly penetrant E200K PRNP mutation and generated a library of induced pluripotent stem cells (iPSCs) representing nine carriers and four non-carriers. iPSC-derived neurons from E200K carriers display abnormal synaptic architecture characterized by misalignment of postsynaptic NMDA receptors with the cytoplasmic scaffolding protein PSD95. Differentiated neurons from mutation carriers do not produce PrPSc, the aggregated and infectious conformer of PrP, suggesting that loss of a physiological function of PrPC may contribute to the disease phenotype. Our study shows that iPSC-derived neurons can provide important mechanistic insights into the pathogenesis of genetic prion diseases and can offer a powerful platform for testing candidate therapeutics.
Collapse
Affiliation(s)
- Aldana D Gojanovich
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
| | - Nhat T T Le
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Robert C C Mercer
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Seonmi Park
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
| | - Bei Wu
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Alice Anane
- Creutzfeldt-Jakob Disease Foundation, Pardes Hanna-Karkur, Israel
| | - Janelle S Vultaggio
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Gustavo Mostoslavsky
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA; Department of Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - David A Harris
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
4
|
Mercer RCC, Harris DA. Mechanisms of prion-induced toxicity. Cell Tissue Res 2022; 392:81-96. [PMID: 36070155 DOI: 10.1007/s00441-022-03683-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022]
Abstract
Prion diseases are devastating neurodegenerative diseases caused by the structural conversion of the normally benign prion protein (PrPC) to an infectious, disease-associated, conformer, PrPSc. After decades of intense research, much is known about the self-templated prion conversion process, a phenomenon which is now understood to be operative in other more common neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. In this review, we provide the current state of knowledge concerning a relatively poorly understood aspect of prion diseases: mechanisms of neurotoxicity. We provide an overview of proposed functions of PrPC and its interactions with other extracellular proteins in the central nervous system, in vivo and in vitro models used to delineate signaling events downstream of prion propagation, the application of omics technologies, and the emerging appreciation of the role played by non-neuronal cell types in pathogenesis.
Collapse
Affiliation(s)
- Robert C C Mercer
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
5
|
Crestini A, Santilli F, Martellucci S, Carbone E, Sorice M, Piscopo P, Mattei V. Prions and Neurodegenerative Diseases: A Focus on Alzheimer's Disease. J Alzheimers Dis 2021; 85:503-518. [PMID: 34864675 DOI: 10.3233/jad-215171] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Specific protein misfolding and aggregation are mechanisms underlying various neurodegenerative diseases such as prion disease and Alzheimer's disease (AD). The misfolded proteins are involved in prions, amyloid-β (Aβ), tau, and α-synuclein disorders; they share common structural, biological, and biochemical characteristics, as well as similar mechanisms of aggregation and self-propagation. Pathological features of AD include the appearance of plaques consisting of deposition of protein Aβ and neurofibrillary tangles formed by the hyperphosphorylated tau protein. Although it is not clear how protein aggregation leads to AD, we are learning that the cellular prion protein (PrPC) plays an important role in the pathogenesis of AD. Herein, we first examined the pathogenesis of prion and AD with a focus on the contribution of PrPC to the development of AD. We analyzed the mechanisms that lead to the formation of a high affinity bond between Aβ oligomers (AβOs) and PrPC. Also, we studied the role of PrPC as an AβO receptor that initiates an AβO-induced signal cascade involving mGluR5, Fyn, Pyk2, and eEF2K linking Aβ and tau pathologies, resulting in the death of neurons in the central nervous system. Finally, we have described how the PrPC-AβOs interaction can be used as a new potential therapeutic target for the treatment of PrPC-dependent AD.
Collapse
Affiliation(s)
- Alessio Crestini
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Santilli
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Rieti, Italy.,Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| | - Stefano Martellucci
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Rieti, Italy
| | - Elena Carbone
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| | - Paola Piscopo
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Rieti, Italy.,Department of Experimental Medicine, "Sapienza" University, Rome, Italy
| |
Collapse
|
6
|
Zanier ER, Barzago MM, Vegliante G, Romeo M, Restelli E, Bertani I, Natale C, Colnaghi L, Colombo L, Russo L, Micotti E, Fioriti L, Chiesa R, Diomede L. C. elegans detects toxicity of traumatic brain injury generated tau. Neurobiol Dis 2021; 153:105330. [PMID: 33711491 PMCID: PMC8039186 DOI: 10.1016/j.nbd.2021.105330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with widespread tau pathology in about 30% of patients surviving late after injury. We previously found that TBI in mice induces the formation of an abnormal form of tau (tauTBI) which progressively spreads from the site of injury to remote brain regions. Intracerebral inoculation of TBI brain homogenates into naïve mice induced progressive tau pathology, synaptic loss and late cognitive decline, suggesting a pivotal role of tauTBI in post-TBI neurodegeneration. However, the possibility that tauTBI was a marker of TBI-associated neurodegeneration rather than a toxic driver of functional decline could not be excluded. Here we employed the nematode C. elegans as a biosensor to test the pathogenic role of TBI generated tau. The motility of this nematode depends on efficient neuromuscular transmission and is exceptionally sensitive to the toxicity of amyloidogenic proteins, providing a tractable model for our tests. We found that worms exposed to brain homogenates from chronic but not acute TBI mice, or from mice in which tauTBI had been transmitted by intracerebral inoculation, had impaired motility and neuromuscular synaptic transmission. Results were similar when worms were given brain homogenates from transgenic mice overexpressing tau P301L, a tauopathy mouse model, suggesting that TBI-induced and mutant tau have similar toxic properties. P301L brain homogenate toxicity was similar in wild-type and ptl-1 knock-out worms, indicating that the nematode tau homolog protein PTL-1 was not required to mediate the toxic effect. Harsh protease digestion to eliminate the protein component of the homogenates, pre-incubation with anti-tau antibodies or tau depletion by immunoprecipitation, abolished the toxicity. Homogenates of chronic TBI brains from tau knock-out mice were not toxic to C. elegans, whereas oligomeric recombinant tau was sufficient to impair their motility. This study indicates that tauTBI impairs motor activity and synaptic transmission in C. elegans and supports a pathogenic role of tauTBI in the long-term consequences of TBI. It also sets the groundwork for the development of a C. elegans-based platform for screening anti-tau compounds. Traumatic brain injury (TBI) in mice induces a progressive tau pathology. Brain-injured tissue from chronic but not acute TBI mice impairs C. elegans motility. TBI tissue immunodepleted of tau or from tau knock-out mice has no toxic effect. Brain-injured tissue from TBI mice impairs neuromuscular transmission in worms. C. elegans is a tractable model for investigating tau toxicity generated by TBI.
Collapse
Affiliation(s)
- Elisa R Zanier
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Maria Monica Barzago
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gloria Vegliante
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Margherita Romeo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elena Restelli
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilaria Bertani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmina Natale
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luca Colnaghi
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luca Russo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luana Fioriti
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Roberto Chiesa
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| |
Collapse
|
7
|
Restelli E, Capone V, Pozzoli M, Ortolan D, Quaglio E, Corbelli A, Fiordaliso F, Beznoussenko GV, Artuso V, Roiter I, Sallese M, Chiesa R. Activation of Src family kinase ameliorates secretory trafficking in mutant prion protein cells. J Biol Chem 2021; 296:100490. [PMID: 33662396 PMCID: PMC8059059 DOI: 10.1016/j.jbc.2021.100490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 02/04/2021] [Accepted: 02/26/2021] [Indexed: 11/25/2022] Open
Abstract
Fatal familial insomnia (FFI), genetic Creutzfeldt-Jakob disease (gCJD), and Gerstmann-Sträussler-Scheinker (GSS) syndrome are neurodegenerative disorders linked to prion protein (PrP) mutations. The pathogenic mechanisms are not known, but increasing evidence points to mutant PrP misfolding and retention in the secretory pathway. We previously found that the D178N/M129 mutation associated with FFI accumulates in the Golgi of neuronal cells, impairing post-Golgi trafficking. In this study we further characterized the trafficking defect induced by the FFI mutation and tested the 178N/V129 variant linked to gCJD and a nine-octapeptide repeat insertion associated with GSS. We used transfected HeLa cells, embryonic fibroblasts and primary neurons from transgenic mice, and fibroblasts from carriers of the FFI mutation. In all these cell types, the mutant PrPs showed abnormal intracellular localizations, accumulating in the endoplasmic reticulum (ER) and Golgi. To test the efficiency of the membrane trafficking system, we monitored the intracellular transport of the temperature-sensitive vesicular stomatite virus glycoprotein (VSV-G), a well-established cargo reporter, and of endogenous procollagen I (PC-I). We observed marked alterations in secretory trafficking, with VSV-G accumulating mainly in the Golgi complex and PC-I in the ER and Golgi. A redacted version of mutant PrP with reduced propensity to misfold did not impair VSV-G trafficking, nor did artificial ER or Golgi retention of wild-type PrP; this indicates that both misfolding and intracellular retention were required to induce the transport defect. Pharmacological activation of Src family kinase (SFK) improved intracellular transport, suggesting that mutant PrP impairs secretory trafficking through corruption of SFK-mediated signaling.
Collapse
Affiliation(s)
- Elena Restelli
- Laboratory of Prion Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Vanessa Capone
- Department of Innovative Technologies in Medicine & Dentistry, University G. D'Annunzio, Chieti, Italy; Center for Advanced Studies and Technology (CAST), University G. D'Annunzio, Chieti, Italy
| | - Manuela Pozzoli
- Laboratory of Prion Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Davide Ortolan
- Laboratory of Prion Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elena Quaglio
- Laboratory of Prion Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Alessandro Corbelli
- Bio-Imaging Unit, Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Fabio Fiordaliso
- Bio-Imaging Unit, Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | - Ignazio Roiter
- ULSS 2 Marca Trevigiana, Ca' Foncello Hospital, Treviso, Italy
| | - Michele Sallese
- Department of Innovative Technologies in Medicine & Dentistry, University G. D'Annunzio, Chieti, Italy; Center for Advanced Studies and Technology (CAST), University G. D'Annunzio, Chieti, Italy
| | - Roberto Chiesa
- Laboratory of Prion Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| |
Collapse
|
8
|
Marín-Moreno A, Espinosa JC, Torres JM. Transgenic mouse models for the study of prion diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:147-177. [PMID: 32958231 DOI: 10.1016/bs.pmbts.2020.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Prions are unique agents that challenge the molecular biology dogma by transmitting information on the protein level. They cause neurodegenerative diseases that lack of any cure or treatment called transmissible spongiform encephalopathies. The function of the normal form of the prion protein, the exact mechanism of prion propagation between species as well as at the cellular level and neuron degeneration remains elusive. However, great amount of information known for all these aspects has been achieved thanks to the use of animal models and more precisely to transgenic mouse models. In this chapter, the main contributions of these powerful research tools in the prion field are revised.
Collapse
Affiliation(s)
- Alba Marín-Moreno
- Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Spain
| | | | - Juan María Torres
- Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Spain.
| |
Collapse
|
9
|
Spagnolli G, Requena JR, Biasini E. Understanding prion structure and conversion. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:19-30. [PMID: 32958233 DOI: 10.1016/bs.pmbts.2020.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Since their original identification, prions have represented enigmatic agents that defy the classical concept of genetic inheritance. For almost four decades, the high-resolution structure of PrPSc, the infectious and misfolded counterpart of the cellular prion protein (PrPC), has remained elusive, mostly due to technical challenges posed by its high insolubility and aggregation propensity. As a result, such a lack of information has critically hampered the search for an effective therapy against prion diseases. Nevertheless, multiple attempts to get insights into the structure of PrPSc have provided important experimental constraints that, despite being at limited resolution, are paving the way for the application of computer-aided technologies to model the three-dimensional architecture of prions and their templated replication mechanism. Here, we review the most relevant studies carried out so far to elucidate the conformation of infectious PrPSc and offer an overview of the most advanced molecular models to explain prion structure and conversion.
Collapse
Affiliation(s)
- Giovanni Spagnolli
- Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, TN, Italy; Dulbecco Telethon Institute, University of Trento, Trento, TN, Italy
| | - Jesús R Requena
- CIMUS Biomedical Research Institute & Department of Medical Sciences, University of Santiago de Compostela-IDIS, Santiago, Spain
| | - Emiliano Biasini
- Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, TN, Italy; Dulbecco Telethon Institute, University of Trento, Trento, TN, Italy.
| |
Collapse
|
10
|
Mutant prion proteins increase calcium permeability of AMPA receptors, exacerbating excitotoxicity. PLoS Pathog 2020; 16:e1008654. [PMID: 32673372 PMCID: PMC7365390 DOI: 10.1371/journal.ppat.1008654] [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: 01/01/2020] [Accepted: 05/26/2020] [Indexed: 01/26/2023] Open
Abstract
Prion protein (PrP) mutations are linked to genetic prion diseases, a class of phenotypically heterogeneous neurodegenerative disorders with invariably fatal outcome. How mutant PrP triggers neurodegeneration is not known. Synaptic dysfunction precedes neuronal loss but it is not clear whether, and through which mechanisms, disruption of synaptic activity ultimately leads to neuronal death. Here we show that mutant PrP impairs the secretory trafficking of AMPA receptors (AMPARs). Specifically, intracellular retention of the GluA2 subunit results in synaptic exposure of GluA2-lacking, calcium-permeable AMPARs, leading to increased calcium permeability and enhanced sensitivity to excitotoxic cell death. Mutant PrPs linked to different genetic prion diseases affect AMPAR trafficking and function in different ways. Our findings identify AMPARs as pathogenic targets in genetic prion diseases, and support the involvement of excitotoxicity in neurodegeneration. They also suggest a mechanistic explanation for how different mutant PrPs may cause distinct disease phenotypes. Genetic prion diseases are degenerative brain disorders caused by mutations in the gene encoding the prion protein (PrP). Different PrP mutations cause different diseases, including Creutzfeldt-Jakob disease, fatal familial insomnia and Gerstmann-Sträussler-Scheinker syndrome. How mutant PrP causes neuronal death and how different mutants encode distinct disease phenotypes is not known. Here we show that mutant PrP alters the subunit composition of glutamate AMPA receptors, promoting cell surface exposure of GluA2-lacking, calcium-permeable receptors, ultimately increasing neuronal vulnerability to excitotoxic cell death. We also demonstrate that the underlying molecular mechanism is the formation of a GluA2 subunit-PrP complex which is retained in the neuronal secretory pathway. PrP mutants associated with clinically different genetic prion diseases have distinct effects on GluA2 trafficking, depending on their tendency to misfold and aggregate in different intracellular organelles, indicating a possible contribution of this mechanism to the disease phenotype.
Collapse
|
11
|
Structural Consequences of Copper Binding to the Prion Protein. Cells 2019; 8:cells8080770. [PMID: 31349611 PMCID: PMC6721516 DOI: 10.3390/cells8080770] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/24/2022] Open
Abstract
Prion, or PrPSc, is the pathological isoform of the cellular prion protein (PrPC) and it is the etiological agent of transmissible spongiform encephalopathies (TSE) affecting humans and animal species. The most relevant function of PrPC is its ability to bind copper ions through its flexible N-terminal moiety. This review includes an overview of the structure and function of PrPC with a focus on its ability to bind copper ions. The state-of-the-art of the role of copper in both PrPC physiology and in prion pathogenesis is also discussed. Finally, we describe the structural consequences of copper binding to the PrPC structure.
Collapse
|
12
|
Forloni G, Chiesa R, Bugiani O, Salmona M, Tagliavini F. Review: PrP 106-126 - 25 years after. Neuropathol Appl Neurobiol 2019; 45:430-440. [PMID: 30635947 DOI: 10.1111/nan.12538] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022]
Abstract
A quarter of a century ago, we proposed an innovative approach to study the pathogenesis of prion disease, one of the most intriguing biomedical problems that remains unresolved. The synthesis of a peptide homologous to residues 106-126 of the human prion protein (PrP106-126), a sequence present in the PrP amyloid protein of Gerstmann-Sträussler-Scheinker syndrome patients, provided a tractable tool for investigating the mechanisms of neurotoxicity. Together with several other discoveries at the beginning of the 1990s, PrP106-126 contributed to underpin the role of amyloid in the pathogenesis of protein-misfolding neurodegenerative disorders. Later, the role of oligomers on one hand and of prion-like spreading of pathology on the other further clarified mechanisms shared by different neurodegenerative conditions. Our original report on PrP106-126 neurotoxicity also highlighted a role for programmed cell death in CNS diseases. In this review, we analyse the prion research context in which PrP106-126 first appeared and the advances in our understanding of prion disease pathogenesis and therapeutic perspectives 25 years later.
Collapse
Affiliation(s)
- G Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - R Chiesa
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - O Bugiani
- Department of Biochemistry, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - M Salmona
- Department of Biochemistry, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - F Tagliavini
- Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milano, Italy
| |
Collapse
|
13
|
Le NTT, Wu B, Harris DA. Prion neurotoxicity. Brain Pathol 2019; 29:263-277. [PMID: 30588688 DOI: 10.1111/bpa.12694] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/28/2018] [Indexed: 01/04/2023] Open
Abstract
Although the mechanisms underlying prion propagation and infectivity are now well established, the processes accounting for prion toxicity and pathogenesis have remained mysterious. These processes are of enormous clinical relevance as they hold the key to identification of new molecular targets for therapeutic intervention. In this review, we will discuss two broad areas of investigation relevant to understanding prion neurotoxicity. The first is the use of in vitro experimental systems that model key events in prion pathogenesis. In this context, we will describe a hippocampal neuronal culture system we developed that reproduces the earliest pathological alterations in synaptic morphology and function in response to PrPSc . This system has allowed us to define a core synaptotoxic signaling pathway involving the activation of NMDA and AMPA receptors, stimulation of p38 MAPK phosphorylation and collapse of the actin cytoskeleton in dendritic spines. The second area concerns a striking and unexpected phenomenon in which certain structural manipulations of the PrPC molecule itself, including introduction of N-terminal deletion mutations or binding of antibodies to C-terminal epitopes, unleash powerful toxic effects in cultured cells and transgenic mice. We will describe our studies of this phenomenon, which led to the recognition that it is related to the induction of large, abnormal ionic currents by the structurally altered PrP molecules. Our results suggest a model in which the flexible N-terminal domain of PrPC serves as a toxic effector which is regulated by intramolecular interactions with the globular C-terminal domain. Taken together, these two areas of study have provided important clues to underlying cellular and molecular mechanisms of prion neurotoxicity. Nevertheless, much remains to be done on this next frontier of prion science.
Collapse
Affiliation(s)
- Nhat T T Le
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Bei Wu
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| |
Collapse
|
14
|
Piccardo P, Asher DM. Complex proteinopathies and neurodegeneration: insights from the study of transmissible spongiform encephalopathies. ARQUIVOS DE NEURO-PSIQUIATRIA 2018; 76:705-712. [PMID: 30427511 DOI: 10.1590/0004-282x20180111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/08/2018] [Indexed: 11/22/2022]
Abstract
Protein misfolding diseases are usually associated with deposits of single "key" proteins that somehow drive the pathology; β-amyloid and hyperphosphorylated tau accumulate in Alzheimer's disease, α-synuclein in Parkinson's disease, or abnormal prion protein (PrPTSE) in transmissible spongiform encephalopathies (TSEs or prion diseases). However, in some diseases more than two proteins accumulate in the same brain. These diseases might be considered "complex" proteinopathies. We have studied models of TSEs (to explore deposits of PrPTSE and of "secondary proteins") infecting different strains and doses of TSE agent, factors that control incubation period, duration of illness and histopathology. Model TSEs allowed us to investigate whether different features of histopathology are independent of PrPTSE or appear as a secondary result of PrPTSE. Better understanding the complex proteinopathies may help to explain the wide spectrum of degenerative diseases and why some overlap clinically and histopathologically. These studies might also improve diagnosis and eventually even suggest new treatments for human neurodegenerative diseases.
Collapse
Affiliation(s)
- Pedro Piccardo
- Laboratory of Bacterial and Transmissible Spongiform Encephalopathy Agents, Division of Emerging and Transfusion-Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration
| | - David M Asher
- Laboratory of Bacterial and Transmissible Spongiform Encephalopathy Agents, Division of Emerging and Transfusion-Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration
| |
Collapse
|
15
|
Abstract
During the course of prion infection, the normally soluble and protease-sensitive mammalian prion protein (PrPC) is refolded into an insoluble, partially protease-resistant, and infectious form called PrPSc. The conformational conversion of PrPC to PrPSc is a critical event during prion infection and is essential for the production of prion infectivity. This chapter briefly summarizes the ways in which cell biological approaches have enhanced our understanding of how PrP contributes to different aspects of prion pathogenesis.
Collapse
|
16
|
Villar-Piqué A, Schmitz M, Candelise N, Ventura S, Llorens F, Zerr I. Molecular and Clinical Aspects of Protein Aggregation Assays in Neurodegenerative Diseases. Mol Neurobiol 2018; 55:7588-7605. [DOI: 10.1007/s12035-018-0926-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 01/24/2018] [Indexed: 12/20/2022]
|
17
|
Prion Protein Devoid of the Octapeptide Repeat Region Delays Bovine Spongiform Encephalopathy Pathogenesis in Mice. J Virol 2017; 92:JVI.01368-17. [PMID: 29046443 DOI: 10.1128/jvi.01368-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/05/2017] [Indexed: 12/26/2022] Open
Abstract
Conformational conversion of the cellular isoform of prion protein, PrPC, into the abnormally folded, amyloidogenic isoform, PrPSc, is a key pathogenic event in prion diseases, including Creutzfeldt-Jakob disease in humans and scrapie and bovine spongiform encephalopathy (BSE) in animals. We previously reported that the octapeptide repeat (OR) region could be dispensable for converting PrPC into PrPSc after infection with RML prions. We demonstrated that mice transgenically expressing mouse PrP with deletion of the OR region on the PrP knockout background, designated Tg(PrPΔOR)/Prnp0/0 mice, did not show reduced susceptibility to RML scrapie prions, with abundant accumulation of PrPScΔOR in their brains. We show here that Tg(PrPΔOR)/Prnp0/0 mice were highly resistant to BSE prions, developing the disease with markedly elongated incubation times after infection with BSE prions. The conversion of PrPΔOR into PrPScΔOR was markedly delayed in their brains. These results suggest that the OR region may have a crucial role in the conversion of PrPC into PrPSc after infection with BSE prions. However, Tg(PrPΔOR)/Prnp0/0 mice remained susceptible to RML and 22L scrapie prions, developing the disease without elongated incubation times after infection with RML and 22L prions. PrPScΔOR accumulated only slightly less in the brains of RML- or 22L-infected Tg(PrPΔOR)/Prnp0/0 mice than PrPSc in control wild-type mice. Taken together, these results indicate that the OR region of PrPC could play a differential role in the pathogenesis of BSE prions and RML or 22L scrapie prions.IMPORTANCE Structure-function relationship studies of PrPC conformational conversion into PrPSc are worthwhile to understand the mechanism of the conversion of PrPC into PrPSc We show here that, by inoculating Tg(PrPΔOR)/Prnp0/0 mice with the three different strains of RML, 22L, and BSE prions, the OR region could play a differential role in the conversion of PrPC into PrPSc after infection with RML or 22L scrapie prions and BSE prions. PrPΔOR was efficiently converted into PrPScΔOR after infection with RML and 22L prions. However, the conversion of PrPΔOR into PrPScΔOR was markedly delayed after infection with BSE prions. Further investigation into the role of the OR region in the conversion of PrPC into PrPSc after infection with BSE prions might be helpful for understanding the pathogenesis of BSE prions.
Collapse
|
18
|
Nonno R, Angelo Di Bari M, Agrimi U, Pirisinu L. Transmissibility of Gerstmann-Sträussler-Scheinker syndrome in rodent models: New insights into the molecular underpinnings of prion infectivity. Prion 2017; 10:421-433. [PMID: 27892798 PMCID: PMC5161296 DOI: 10.1080/19336896.2016.1239686] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Prion diseases, or transmissible spongiform encephalopathies, have revealed the bewildering phenomenon of transmissibility in neurodegenerative diseases. Hence, the experimental transmissibility of prion-like neurodegenerative diseases via template directed misfolding has become the focus of intense research. Gerstmann-Sträussler-Scheinker disease (GSS) is an inherited prion disease associated with mutations in the prion protein gene. However, with the exception of a few GSS cases with P102L mutation characterized by co-accumulation of protease-resistant PrP core (PrPres) of ∼21 kDa, attempts to transmit to rodents GSS associated to atypical misfolded prion protein with ∼8 kDa PrPres have been unsuccessful. As a result, these GSS subtypes have often been considered as non-transmissible proteinopathies rather than true prion diseases. In a recent study we inoculated bank voles with GSS cases associated with P102L, A117V and F198S mutations and found that they transmitted efficiently and produced distinct pathological phenotypes, irrespective of the presence of 21 kDa PrPres in the inoculum. This study demonstrates that GSS is a genuine prion disease characterized by both transmissibility and strain variation. We discuss the implications of these findings for the understanding of the heterogeneous clinic-pathological phenotypes of GSS and of the molecular underpinnings of prion infectivity.
Collapse
Affiliation(s)
- Romolo Nonno
- a Department of Veterinary Public Health and Food Safety , Istituto Superiore di Sanità , Rome , Italy
| | - Michele Angelo Di Bari
- a Department of Veterinary Public Health and Food Safety , Istituto Superiore di Sanità , Rome , Italy
| | - Umberto Agrimi
- a Department of Veterinary Public Health and Food Safety , Istituto Superiore di Sanità , Rome , Italy
| | - Laura Pirisinu
- a Department of Veterinary Public Health and Food Safety , Istituto Superiore di Sanità , Rome , Italy
| |
Collapse
|
19
|
Chiesa R, Restelli E, Comerio L, Del Gallo F, Imeri L. Transgenic mice recapitulate the phenotypic heterogeneity of genetic prion diseases without developing prion infectivity: Role of intracellular PrP retention in neurotoxicity. Prion 2017; 10:93-102. [PMID: 26864450 PMCID: PMC4981194 DOI: 10.1080/19336896.2016.1139276] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Genetic prion diseases are degenerative brain disorders caused by mutations in the gene encoding the prion protein (PrP). Different PrP mutations cause different diseases, including Creutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker (GSS) syndrome and fatal familial insomnia (FFI). The reason for this variability is not known. It has been suggested that prion strains with unique self-replicating and neurotoxic properties emerge spontaneously in individuals carrying PrP mutations, dictating the phenotypic expression of disease. We generated transgenic mice expressing the FFI mutation, and found that they developed a fatal neurological illness highly reminiscent of FFI, and different from those of similarly generated mice modeling genetic CJD and GSS. Thus transgenic mice recapitulate the phenotypic differences seen in humans. The mutant PrPs expressed in these mice are misfolded but unable to self-replicate. They accumulate in different compartments of the neuronal secretory pathway, impairing the membrane delivery of ion channels essential for neuronal function. Our results indicate that conversion of mutant PrP into an infectious isoform is not required for pathogenesis, and suggest that the phenotypic variability may be due to different effects of mutant PrP on intracellular transport.
Collapse
Affiliation(s)
- Roberto Chiesa
- a Laboratory of Prion Neurobiology, Department of Neuroscience, IRCCS - "Mario Negri" Institute for Pharmacological Research , Milan , Italy
| | - Elena Restelli
- a Laboratory of Prion Neurobiology, Department of Neuroscience, IRCCS - "Mario Negri" Institute for Pharmacological Research , Milan , Italy
| | - Liliana Comerio
- a Laboratory of Prion Neurobiology, Department of Neuroscience, IRCCS - "Mario Negri" Institute for Pharmacological Research , Milan , Italy
| | - Federico Del Gallo
- b Department of Health Sciences , University of Milan Medical School , Milan , Italy
| | - Luca Imeri
- b Department of Health Sciences , University of Milan Medical School , Milan , Italy
| |
Collapse
|
20
|
Comoy EE, Mikol J, Jaffré N, Lebon V, Levavasseur E, Streichenberger N, Sumian C, Perret-Liaudet A, Eloit M, Andreoletti O, Haïk S, Hantraye P, Deslys JP. Experimental transfusion of variant CJD-infected blood reveals previously uncharacterised prion disorder in mice and macaque. Nat Commun 2017; 8:1268. [PMID: 29097653 PMCID: PMC5668246 DOI: 10.1038/s41467-017-01347-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 09/11/2017] [Indexed: 12/12/2022] Open
Abstract
Exposure of human populations to bovine spongiform encephalopathy through contaminated food has resulted in <250 cases of variant Creutzfeldt-Jakob disease (vCJD). However, more than 99% of vCJD infections could have remained silent suggesting a long-term risk of secondary transmission particularly through blood. Here, we present experimental evidence that transfusion in mice and non-human primates of blood products from symptomatic and non-symptomatic infected donors induces not only vCJD, but also a different class of neurological impairments. These impairments can all be retransmitted to mice with a pathognomonic accumulation of abnormal prion protein, thus expanding the spectrum of known prion diseases. Our findings suggest that the intravenous route promotes propagation of masked prion variants according to different mechanisms involved in peripheral replication.
Collapse
Affiliation(s)
- Emmanuel E Comoy
- CEA, Institut François Jacob, Université Paris-Saclay, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France.
| | - Jacqueline Mikol
- CEA, Institut François Jacob, Université Paris-Saclay, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Nina Jaffré
- CEA, Institut François Jacob, Université Paris-Saclay, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
- MacoPharma, 200 Chaussée Fernand Forest, 59200, Tourcoing, France
| | - Vincent Lebon
- CEA, Institut François Jacob, Université Paris-Saclay, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Etienne Levavasseur
- Université Pierre et Marie Curie, UMR-S 1127, CNRS UMR 722, Institut du Cerveau et de la Moelle Epinière, G.H. Pitié-Salpêtrière, 47 Boulevard de l'Hôpital, 75013, Paris, France
| | - Nathalie Streichenberger
- Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyogène CNRS UMR 5310-INSERM U1217, 59 Boulevard Pinel, 69677, Bron, France
| | - Chryslain Sumian
- MacoPharma, 200 Chaussée Fernand Forest, 59200, Tourcoing, France
| | - Armand Perret-Liaudet
- Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyogène CNRS UMR 5310-INSERM U1217, 59 Boulevard Pinel, 69677, Bron, France
| | - Marc Eloit
- Institut Pasteur, 15 Rue du Docteur Roux, 75015, Paris, France
| | - Olivier Andreoletti
- UMR INRA-ENVT 1225, Ecole Nationale Vétérinaire de Toulouse, 23 chemin des Capelles, 31076, Toulouse, France
| | - Stéphane Haïk
- Université Pierre et Marie Curie, UMR-S 1127, CNRS UMR 722, Institut du Cerveau et de la Moelle Epinière, G.H. Pitié-Salpêtrière, 47 Boulevard de l'Hôpital, 75013, Paris, France
| | - Philippe Hantraye
- CEA, Institut François Jacob, Université Paris-Saclay, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| | - Jean-Philippe Deslys
- CEA, Institut François Jacob, Université Paris-Saclay, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
| |
Collapse
|
21
|
Watts JC, Prusiner SB. Experimental Models of Inherited PrP Prion Diseases. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a027151. [PMID: 28096244 DOI: 10.1101/cshperspect.a027151] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The inherited prion protein (PrP) prion disorders, which include familial Creutzfeldt-Jakob disease, Gerstmann-Sträussler-Scheinker disease, and fatal familial insomnia, constitute ∼10%-15% of all PrP prion disease cases in humans. Attempts to generate animal models of these disorders using transgenic mice expressing mutant PrP have produced variable results. Although many lines of mice develop spontaneous signs of neurological illness with accompanying prion disease-specific neuropathological changes, others do not. Furthermore, demonstrating the presence of protease-resistant PrP species and prion infectivity-two of the hallmarks of the PrP prion disorders-in the brains of spontaneously sick mice has proven particularly challenging. Here, we review the progress that has been made toward developing accurate mouse models of the inherited PrP prion disorders.
Collapse
Affiliation(s)
- Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Biochemistry, University of Toronto, Toronto, Ontario M5T 2S8, Canada
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, Departments of Neurology and Biochemistry and Biophysics, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
22
|
Ke PC, Sani MA, Ding F, Kakinen A, Javed I, Separovic F, Davis TP, Mezzenga R. Implications of peptide assemblies in amyloid diseases. Chem Soc Rev 2017; 46:6492-6531. [PMID: 28702523 PMCID: PMC5902192 DOI: 10.1039/c7cs00372b] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neurodegenerative disorders and type 2 diabetes are global epidemics compromising the quality of life of millions worldwide, with profound social and economic implications. Despite the significant differences in pathology - much of which are poorly understood - these diseases are commonly characterized by the presence of cross-β amyloid fibrils as well as the loss of neuronal or pancreatic β-cells. In this review, we document research progress on the molecular and mesoscopic self-assembly of amyloid-beta, alpha synuclein, human islet amyloid polypeptide and prions, the peptides and proteins associated with Alzheimer's, Parkinson's, type 2 diabetes and prion diseases. In addition, we discuss the toxicities of these amyloid proteins based on their self-assembly as well as their interactions with membranes, metal ions, small molecules and engineered nanoparticles. Through this presentation we show the remarkable similarities and differences in the structural transitions of the amyloid proteins through primary and secondary nucleation, the common evolution from disordered monomers to alpha-helices and then to β-sheets when the proteins encounter the cell membrane, and, the consensus (with a few exceptions) that off-pathway oligomers, rather than amyloid fibrils, are the toxic species regardless of the pathogenic protein sequence or physicochemical properties. In addition, we highlight the crucial role of molecular self-assembly in eliciting the biological and pathological consequences of the amyloid proteins within the context of their cellular environments and their spreading between cells and organs. Exploiting such structure-function-toxicity relationship may prove pivotal for the detection and mitigation of amyloid diseases.
Collapse
Affiliation(s)
- Pu Chun Ke
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Marc-Antonie Sani
- School of Chemistry, Bio21 Institute, The University of Melbourne, 30 Flemington Rd, Parkville, VIC 3010, Australia
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Aleksandr Kakinen
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Ibrahim Javed
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Frances Separovic
- School of Chemistry, Bio21 Institute, The University of Melbourne, 30 Flemington Rd, Parkville, VIC 3010, Australia
| | - Thomas P. Davis
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Department of Chemistry, University of Warwick, Gibbet Hill, Coventry, CV4 7AL, United Kingdom
| | - Raffaele Mezzenga
- ETH Zurich, Department of Health Science & Technology, Schmelzbergstrasse 9, LFO, E23, 8092 Zurich, Switzerland
| |
Collapse
|
23
|
Moreno JA, Telling GC. Insights into Mechanisms of Transmission and Pathogenesis from Transgenic Mouse Models of Prion Diseases. Methods Mol Biol 2017; 1658:219-252. [PMID: 28861793 DOI: 10.1007/978-1-4939-7244-9_16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Prions represent a new paradigm of protein-mediated information transfer. In the case of mammals, prions are the cause of fatal, transmissible neurodegenerative diseases, sometimes referred to as transmissible spongiform encephalopathies (TSEs), which frequently occur as epidemics. An increasing body of evidence indicates that the canonical mechanism of conformational corruption of cellular prion protein (PrPC) by the pathogenic isoform (PrPSc) that is the basis of prion formation in TSEs is common to a spectrum of proteins associated with various additional human neurodegenerative disorders, including the more common Alzheimer's and Parkinson's diseases. The peerless infectious properties of TSE prions, and the unparalleled tools for their study, therefore enable elucidation of mechanisms of template-mediated conformational propagation that are generally applicable to these related disease states. Many unresolved issues remain including the exact molecular nature of the prion, the detailed cellular and molecular mechanisms of prion propagation, and the means by which prion diseases can be both genetic and infectious. In addition, we know little about the mechanism by which neurons degenerate during prion diseases. Tied to this, the physiological role of the normal form of the prion protein remains unclear and it is uncertain whether or not loss of this function contributes to prion pathogenesis. The factors governing the transmission of prions between species remain unclear, in particular the means by which prion strains and PrP primary structure interact to affect interspecies prion transmission. Despite all these unknowns, advances in our understanding of prions have occurred because of their transmissibility to experimental animals, and the development of transgenic (Tg) mouse models has done much to further our understanding about various aspects of prion biology. In this review, we will focus on advances in our understanding of prion biology that occurred in the past 8 years since our last review of this topic.
Collapse
Affiliation(s)
- Julie A Moreno
- Cell and Molecular Biology Graduate Program, Molecular, Cellular and Integrative Neuroscience Graduate Program, Department of Microbiology, Immunology and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, 80523, USA
| | - Glenn C Telling
- Cell and Molecular Biology Graduate Program, Molecular, Cellular and Integrative Neuroscience Graduate Program, Department of Microbiology, Immunology and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, 80523, USA.
| |
Collapse
|
24
|
Barron RM, King D, Jeffrey M, McGovern G, Agarwal S, Gill AC, Piccardo P. PrP aggregation can be seeded by pre-formed recombinant PrP amyloid fibrils without the replication of infectious prions. Acta Neuropathol 2016; 132:611-24. [PMID: 27376534 PMCID: PMC5023723 DOI: 10.1007/s00401-016-1594-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/08/2016] [Accepted: 06/26/2016] [Indexed: 12/22/2022]
Abstract
Mammalian prions are unusual infectious agents, as they are thought to consist solely of aggregates of misfolded prion protein (PrP). Generation of synthetic prions, composed of recombinant PrP (recPrP) refolded into fibrils, has been utilised to address whether PrP aggregates are, indeed, infectious prions. In several reports, neurological disease similar to transmissible spongiform encephalopathy (TSE) has been described following inoculation and passage of various forms of fibrils in transgenic mice and hamsters. However, in studies described here, we show that inoculation of recPrP fibrils does not cause TSE disease, but, instead, seeds the formation of PrP amyloid plaques in PrP-P101L knock-in transgenic mice (101LL). Importantly, both WT-recPrP fibrils and 101L-recPrP fibrils can seed plaque formation, indicating that the fibrillar conformation, and not the primary sequence of PrP in the inoculum, is important in initiating seeding. No replication of infectious prions or TSE disease was observed following both primary inoculation and subsequent subpassage. These data, therefore, argue against recPrP fibrils being infectious prions and, instead, indicate that these pre-formed seeds are acting to accelerate the formation of PrP amyloid plaques in 101LL Tg mice. In addition, these data reproduce a phenotype which was previously observed in 101LL mice following inoculation with brain extract containing in vivo-generated PrP amyloid fibrils, which has not been shown for other synthetic prion models. These data are reminiscent of the "prion-like" spread of aggregated forms of the beta-amyloid peptide (Aβ), α-synuclein and tau observed following inoculation of transgenic mice with pre-formed seeds of each misfolded protein. Hence, even when the protein is PrP, misfolding and aggregation do not reproduce the full clinicopathological phenotype of disease. The initiation and spread of protein aggregation in transgenic mouse lines following inoculation with pre-formed fibrils may, therefore, more closely resemble a seeded proteinopathy than an infectious TSE disease.
Collapse
Affiliation(s)
- Rona M Barron
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK.
| | - Declan King
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK
| | - Martin Jeffrey
- Animal and Plant Health Agency, Pentlands Science Park, Midlothian, Scotland, UK
| | - Gillian McGovern
- Animal and Plant Health Agency, Pentlands Science Park, Midlothian, Scotland, UK
| | - Sonya Agarwal
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK
| | - Andrew C Gill
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK
| | - Pedro Piccardo
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK
| |
Collapse
|
25
|
A Neuronal Culture System to Detect Prion Synaptotoxicity. PLoS Pathog 2016; 12:e1005623. [PMID: 27227882 PMCID: PMC4881977 DOI: 10.1371/journal.ppat.1005623] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/19/2016] [Indexed: 12/02/2022] Open
Abstract
Synaptic pathology is an early feature of prion as well as other neurodegenerative diseases. Although the self-templating process by which prions propagate is well established, the mechanisms by which prions cause synaptotoxicity are poorly understood, due largely to the absence of experimentally tractable cell culture models. Here, we report that exposure of cultured hippocampal neurons to PrPSc, the infectious isoform of the prion protein, results in rapid retraction of dendritic spines. This effect is entirely dependent on expression of the cellular prion protein, PrPC, by target neurons, and on the presence of a nine-amino acid, polybasic region at the N-terminus of the PrPC molecule. Both protease-resistant and protease-sensitive forms of PrPSc cause dendritic loss. This system provides new insights into the mechanisms responsible for prion neurotoxicity, and it provides a platform for characterizing different pathogenic forms of PrPSc and testing potential therapeutic agents. Prion diseases are fatal neurodegenerative disorders that cause memory loss, impaired coordination, and abnormal movements. The molecular culprit in prion diseases is PrPSc, an infectious isoform of a host-encoded glycoprotein (PrPC) that can propagate itself by a self-templating mechanism. Whether PrPSc itself is toxic to neurons, and if so, the cellular mechanisms by which it produces neuronal pathology are largely unknown, in part because of the absence of suitable cell culture models. We describe here a hippocampal neuronal cultural system to detect the toxic effect of PrPSc on dendritic spines, which are postsynaptic elements responsible for excitatory synaptic transmission, and which are implicated in learning, memory, and the earliest stages of neurodegenerative diseases. We found that purified, exogenously applied PrPSc causes acute retraction of dendritic spines, an effect that is entirely dependent on expression of PrPC by target neurons, and on the on the presence of a nine-amino acid, polybasic region at the N-terminus of the PrPC molecule. Both protease-resistant and protease-sensitive forms of PrPSc cause dendritic retraction. This system provides new insights into the mechanisms responsible for prion neurotoxicity, and it provides a platform for characterizing different pathogenic forms of PrPSc and testing potential therapeutic agents.
Collapse
|
26
|
Abstract
Transmissible spongiform encephalopathies (TSEs), or prion diseases, are fatal neurodegenerative disorders characterised by long incubation period, short clinical duration, and transmissibility to susceptible species. Neuronal loss, spongiform changes, gliosis and the accumulation in the brain of the misfolded version of a membrane-bound cellular prion protein (PrP(C)), termed PrP(TSE), are diagnostic markers of these diseases. Compelling evidence links protein misfolding and its accumulation with neurodegenerative changes. Accordingly, several mechanisms of prion-mediated neurotoxicity have been proposed. In this paper, we provide an overview of the recent knowledge on the mechanisms of neuropathogenesis, the neurotoxic PrP species and the possible therapeutic approaches to treat these devastating disorders.
Collapse
|
27
|
Imberdis T, Harris DA. Synthetic Prions Provide Clues for Understanding Prion Diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:761-4. [PMID: 26854642 DOI: 10.1016/j.ajpath.2015.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 12/19/2015] [Indexed: 02/02/2023]
Abstract
This Commentary highlights the article by Makarava et al that discusses the formation of synthetic prions and the role of substrate levels in their evolution.
Collapse
Affiliation(s)
- Thibaut Imberdis
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
28
|
Pirisinu L, Di Bari MA, D'Agostino C, Marcon S, Riccardi G, Poleggi A, Cohen ML, Appleby BS, Gambetti P, Ghetti B, Agrimi U, Nonno R. Gerstmann-Sträussler-Scheinker disease subtypes efficiently transmit in bank voles as genuine prion diseases. Sci Rep 2016; 6:20443. [PMID: 26841849 PMCID: PMC4740801 DOI: 10.1038/srep20443] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/04/2016] [Indexed: 11/16/2022] Open
Abstract
Gerstmann-Sträussler-Scheinker disease (GSS) is an inherited neurodegenerative disorder associated with mutations in the prion protein gene and accumulation of misfolded PrP with protease-resistant fragments (PrPres) of 6–8 kDa. With the exception of a few GSS cases characterized by co-accumulation of PrPres of 21 kDa, efforts to transmit GSS to rodents have been unsuccessful. As a result, GSS subtypes exclusively associated with 6–8 kDa PrPres have often been considered as non-transmissible proteinopathies rather than true prion diseases. We show that GSS with P102L, A117V and F198S mutations transmit efficiently and produce distinct pathological phenotypes in bank voles (M. glareolus), irrespective of the presence of 21 kDa PrPres in the inoculum, demonstrating that GSS is a genuine prion disease characterized by both transmissibility and strain variation.
Collapse
Affiliation(s)
- Laura Pirisinu
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Michele A Di Bari
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Claudia D'Agostino
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Stefano Marcon
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Geraldina Riccardi
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Anna Poleggi
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Mark L Cohen
- Department of Pathology, National Prion Disease Pathology Surveillance Center, Case Western Reserve University, School of Medicine, 2085 Adelbert Road Cleveland, Ohio, OH 44106, USA
| | - Brian S Appleby
- Department of Pathology, National Prion Disease Pathology Surveillance Center, Case Western Reserve University, School of Medicine, 2085 Adelbert Road Cleveland, Ohio, OH 44106, USA
| | - Pierluigi Gambetti
- Department of Pathology, National Prion Disease Pathology Surveillance Center, Case Western Reserve University, School of Medicine, 2085 Adelbert Road Cleveland, Ohio, OH 44106, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Umberto Agrimi
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Romolo Nonno
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
29
|
Insights into Mechanisms of Chronic Neurodegeneration. Int J Mol Sci 2016; 17:ijms17010082. [PMID: 26771599 PMCID: PMC4730326 DOI: 10.3390/ijms17010082] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/22/2015] [Accepted: 12/23/2015] [Indexed: 12/03/2022] Open
Abstract
Chronic neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and prion diseases are characterised by the accumulation of abnormal conformers of a host encoded protein in the central nervous system. The process leading to neurodegeneration is still poorly defined and thus development of early intervention strategies is challenging. Unique amongst these diseases are Transmissible Spongiform Encephalopathies (TSEs) or prion diseases, which have the ability to transmit between individuals. The infectious nature of these diseases has permitted in vivo and in vitro modelling of the time course of the disease process in a highly reproducible manner, thus early events can be defined. Recent evidence has demonstrated that the cell-to-cell spread of protein aggregates by a “prion-like mechanism” is common among the protein misfolding diseases. Thus, the TSE models may provide insights into disease mechanisms and testable hypotheses for disease intervention, applicable to a number of these chronic neurodegenerative diseases.
Collapse
|
30
|
Bouybayoune I, Mantovani S, Del Gallo F, Bertani I, Restelli E, Comerio L, Tapella L, Baracchi F, Fernández-Borges N, Mangieri M, Bisighini C, Beznoussenko GV, Paladini A, Balducci C, Micotti E, Forloni G, Castilla J, Fiordaliso F, Tagliavini F, Imeri L, Chiesa R. Transgenic fatal familial insomnia mice indicate prion infectivity-independent mechanisms of pathogenesis and phenotypic expression of disease. PLoS Pathog 2015; 11:e1004796. [PMID: 25880443 PMCID: PMC4400166 DOI: 10.1371/journal.ppat.1004796] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 03/09/2015] [Indexed: 11/18/2022] Open
Abstract
Fatal familial insomnia (FFI) and a genetic form of Creutzfeldt-Jakob disease (CJD178) are clinically different prion disorders linked to the D178N prion protein (PrP) mutation. The disease phenotype is determined by the 129 M/V polymorphism on the mutant allele, which is thought to influence D178N PrP misfolding, leading to the formation of distinctive prion strains with specific neurotoxic properties. However, the mechanism by which misfolded variants of mutant PrP cause different diseases is not known. We generated transgenic (Tg) mice expressing the mouse PrP homolog of the FFI mutation. These mice synthesize a misfolded form of mutant PrP in their brains and develop a neurological illness with severe sleep disruption, highly reminiscent of FFI and different from that of analogously generated Tg(CJD) mice modeling CJD178. No prion infectivity was detectable in Tg(FFI) and Tg(CJD) brains by bioassay or protein misfolding cyclic amplification, indicating that mutant PrP has disease-encoding properties that do not depend on its ability to propagate its misfolded conformation. Tg(FFI) and Tg(CJD) neurons have different patterns of intracellular PrP accumulation associated with distinct morphological abnormalities of the endoplasmic reticulum and Golgi, suggesting that mutation-specific alterations of secretory transport may contribute to the disease phenotype. Genetic prion diseases are degenerative brain disorders caused by mutations in the gene encoding the prion protein (PrP). Different PrP mutations cause different diseases, including Creutzfeldt-Jakob disease (CJD) and fatal familial insomnia (FFI). The reason for this variability is not known, but assembly of the mutant PrPs into distinct aggregates that spread in the brain by promoting PrP aggregation may contribute to the disease phenotype. We previously generated transgenic mice modeling genetic CJD, clinically identified by dementia and motor abnormalities. We have now generated transgenic mice carrying the PrP mutation associated with FFI, and found that they develop severe sleep abnormalities and other key features of the human disorder. Thus, transgenic mice recapitulate the phenotypic differences seen in humans. The mutant PrPs in FFI and CJD mice are aggregated but unable to promote PrP aggregation. They accumulate in different intracellular compartments and cause distinct morphological abnormalities of transport organelles. These results indicate that mutant PrP has disease-encoding properties that are independent of its ability to self-propagate, and suggest that the phenotypic heterogeneity may be due to different effects of aggregated PrP on intracellular transport. Our study provides new insights into the mechanisms of selective neuronal dysfunction due to protein aggregation.
Collapse
Affiliation(s)
- Ihssane Bouybayoune
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Susanna Mantovani
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Federico Del Gallo
- Department of Health Sciences, University of Milan Medical School, Milan, Italy
| | - Ilaria Bertani
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Elena Restelli
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Liliana Comerio
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Laura Tapella
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Francesca Baracchi
- Department of Health Sciences, University of Milan Medical School, Milan, Italy
| | | | - Michela Mangieri
- Division of Neuropathology and Neurology, IRCCS Foundation “Carlo Besta” National Neurological Institute, Milan, Italy
| | - Cinzia Bisighini
- Bio-Imaging Unit, Department of Cardiovascular Research, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | | | - Alessandra Paladini
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Claudia Balducci
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Joaquín Castilla
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Fabio Fiordaliso
- Bio-Imaging Unit, Department of Cardiovascular Research, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Fabrizio Tagliavini
- Division of Neuropathology and Neurology, IRCCS Foundation “Carlo Besta” National Neurological Institute, Milan, Italy
| | - Luca Imeri
- Department of Health Sciences, University of Milan Medical School, Milan, Italy
| | - Roberto Chiesa
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
- * E-mail:
| |
Collapse
|
31
|
Torres JM, Castilla J, Pintado B, Gutiérrez-Adan A, Andréoletti O, Aguilar-Calvo P, Arroba AI, Parra-Arrondo B, Ferrer I, Manzanares J, Espinosa JC. Spontaneous generation of infectious prion disease in transgenic mice. Emerg Infect Dis 2014; 19:1938-47. [PMID: 24274622 PMCID: PMC3840888 DOI: 10.3201/eid1912.130106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We generated transgenic mice expressing bovine cellular prion protein (PrPC) with a leucine substitution at codon 113 (113L). This protein is homologous to human protein with mutation 102L, and its genetic link with Gerstmann–Sträussler–Scheinker syndrome has been established. This mutation in bovine PrPC causes a fully penetrant, lethal, spongiform encephalopathy. This genetic disease was transmitted by intracerebral inoculation of brain homogenate from ill mice expressing mutant bovine PrP to mice expressing wild-type bovine PrP, which indicated de novo generation of infectious prions. Our findings demonstrate that a single amino acid change in the PrPC sequence can induce spontaneous generation of an infectious prion disease that differs from all others identified in hosts expressing the same PrPC sequence. These observations support the view that a variety of infectious prion strains might spontaneously emerge in hosts displaying random genetic PrPC mutations.
Collapse
|
32
|
Davidson L, Knight R. Neuropathogenesis of prion disease. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.13.74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT: Although much is known about prion diseases (characterized by a post-translational misfolding of the prion protein [PrP]) and their neuropathology and molecular pathology, the fundamental cause of illness, the basic neuropathogenesis, remains uncertain. There are three broad considerations discussed in this review: the possible loss of normal PrP function, the possible direct toxicity of the abnormally folded PrP and a harmful interaction between the normal and abnormal protein. In considering these possibilities, there are difficulties, including the facts that the relevant normal functions of the PrP are somewhat uncertain and that there are a number of possible toxic species of abnormal protein. In addition to the possible interactions of normal and abnormal PrP in prion disease, PrP may play a role in the neuropathogenesis of other diseases (such as Alzheimer’s disease).
Collapse
Affiliation(s)
- Louise Davidson
- National Creutzfeldt–Jakob Disease Research & Surveillance Unit, University of Edinburgh, Edinburgh, UK
| | - Richard Knight
- National Creutzfeldt–Jakob Disease Research & Surveillance Unit, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
33
|
Prion protein misfolding, strains, and neurotoxicity: an update from studies on Mammalian prions. Int J Cell Biol 2013; 2013:910314. [PMID: 24454379 PMCID: PMC3884631 DOI: 10.1155/2013/910314] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 11/10/2013] [Accepted: 11/11/2013] [Indexed: 11/17/2022] Open
Abstract
Prion diseases, also known as transmissible spongiform encephalopathies (TSEs), are a group of fatal neurodegenerative disorders affecting humans and other mammalian species. The central event in TSE pathogenesis is the conformational conversion of the cellular prion protein, PrPC, into the aggregate, β-sheet rich, amyloidogenic form, PrPSc. Increasing evidence indicates that distinct PrPSc conformers, forming distinct ordered aggregates, can encipher the phenotypic TSE variants related to prion strains. Prion strains are TSE isolates that, after inoculation into syngenic hosts, cause disease with distinct characteristics, such as incubation period, pattern of PrPSc distribution, and regional severity of histopathological changes in the brain. In analogy with other amyloid forming proteins, PrPSc toxicity is thought to derive from the existence of various intermediate structures prior to the amyloid fiber formation and/or their specific interaction with membranes. The latter appears particularly relevant for the pathogenesis of TSEs associated with GPI-anchored PrPSc, which involves major cellular membrane distortions in neurons. In this review, we update the current knowledge on the molecular mechanisms underlying three fundamental aspects of the basic biology of prions such as the putative mechanism of prion protein conversion to the pathogenic form PrPSc and its propagation, the molecular basis of prion strains, and the mechanism of induced neurotoxicity by PrPSc aggregates.
Collapse
|
34
|
Synaptic dysfunction in prion diseases: a trafficking problem? Int J Cell Biol 2013; 2013:543803. [PMID: 24369467 PMCID: PMC3863542 DOI: 10.1155/2013/543803] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/08/2013] [Indexed: 11/26/2022] Open
Abstract
Synaptic dysfunction is an important cause of neurological symptoms in prion diseases, a class of clinically heterogeneous neurodegenerative disorders caused by misfolding of the cellular prion protein (PrPC). Experimental data suggest that accumulation of misfolded PrPC in the endoplasmic reticulum (ER) may be crucial in synaptic failure, possibly because of the activation of the translational repression pathway of the unfolded protein response. Here, we report that this pathway is not operative in mouse models of genetic prion disease, consistent with our previous observation that ER stress is not involved. Building on our recent finding that ER retention of mutant PrPC impairs the secretory trafficking of calcium channels essential for synaptic function, we propose a model of pathogenicity in which intracellular retention of misfolded PrPC results in loss of function or gain of toxicity of PrPC-interacting proteins. This neurotoxic modality may also explain the phenotypic heterogeneity of prion diseases.
Collapse
|
35
|
Epitope scanning indicates structural differences in brain-derived monomeric and aggregated mutant prion proteins related to genetic prion diseases. Biochem J 2013; 454:417-25. [PMID: 23808898 DOI: 10.1042/bj20130563] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genetic Creutzfeldt-Jakob disease, Gerstmann-Sträussler-Scheinker syndrome, fatal familial insomnia and prion protein cerebral amyloid angiopathy are clinically and neuropathologically distinct neurodegenerative diseases linked to mutations in the PRNP gene encoding the cellular prion protein (PrPC). How sequence variants of PRNP encode the information to specify these disease phenotypes is not known. It is suggested that each mutation produces a misfolded variant of PrPC with specific neurotoxic properties. However, structural studies of recombinant PrP did not detect major differences between wild-type and mutant molecules, pointing to the importance of investigating mutant PrPs from mammalian brains. We used surface plasmon resonance and a slot-blot immunoassay to analyse the antibody-binding profiles of soluble and insoluble PrP molecules extracted from the brains of transgenic mice modelling different prion diseases. By measuring the reactivity of monoclonal antibodies against different PrP epitopes, we obtained evidence of conformational differences between wild-type and mutant PrPs, and among different mutants. We detected structural heterogeneity in both monomeric and aggregated PrP, supporting the hypothesis that the phenotype of genetic prion diseases is encoded by mutant PrP conformation and assembly state.
Collapse
|
36
|
Dissociation of prion protein amyloid seeding from transmission of a spongiform encephalopathy. J Virol 2013; 87:12349-56. [PMID: 24027305 PMCID: PMC3807897 DOI: 10.1128/jvi.00673-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Misfolding and aggregation of proteins are common pathogenic mechanisms of a group of diseases called proteinopathies. The formation and spread of proteinaceous lesions within and between individuals were first described in prion diseases and proposed as the basis of their infectious nature. Recently, a similar "prion-like" mechanism of transmission has been proposed in other neurodegenerative diseases such as Alzheimer's disease. We investigated if misfolding and aggregation of corrupted prion protein (PrP(TSE)) are always associated with horizontal transmission of disease. Knock-in transgenic mice (101LL) expressing mutant PrP (PrP-101L) that are susceptible to disease but do not develop any spontaneous neurological phenotype were inoculated with (i) brain extracts containing PrP(TSE) from healthy 101LL mice with PrP plaques in the corpus callosum or (ii) brain extracts from mice overexpressing PrP-101L with neurological disease, severe spongiform encephalopathy, and formation of proteinase K-resistant PrP(TSE). In all instances, 101LL mice developed PrP plaques in the area of inoculation and vicinity in the absence of clinical disease or spongiform degeneration of the brain. Importantly, 101LL mice did not transmit disease on serial passage, ruling out the presence of subclinical infection. Thus, in both experimental models the formation of PrP(TSE) is not infectious. These results have implications for the interpretation of tests based on the detection of protein aggregates and suggest that de novo formation of PrP(TSE) in the host does not always result in a transmissible prion disease. In addition, these results question the validity of assuming that all diseases due to protein misfolding can be transmitted between individuals.
Collapse
|
37
|
Gendreau KL, Hall GF. Tangles, Toxicity, and Tau Secretion in AD - New Approaches to a Vexing Problem. Front Neurol 2013; 4:160. [PMID: 24151487 PMCID: PMC3801151 DOI: 10.3389/fneur.2013.00160] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/26/2013] [Indexed: 12/14/2022] Open
Abstract
When the microtubule (MT)-associated protein tau is not bound to axonal MTs, it becomes hyperphosphorylated and vulnerable to proteolytic cleavage and other changes typically seen in the hallmark tau deposits (neurofibrillary tangles) of tau-associated neurodegenerative diseases (tauopathies). Neurofibrillary tangle formation is preceded by tau oligomerization and accompanied by covalent crosslinking and cytotoxicity, making tangle cytopathogenesis a natural central focus of studies directed at understanding the role of tau in neurodegenerative disease. Recent studies suggest that the formation of tau oligomers may be more closely related to tau neurotoxicity than the presence of the tangles themselves. It has also become increasingly clear that tau pathobiology involves a wide variety of other cellular abnormalities including a disruption of autophagy, vesicle trafficking mechanisms, axoplasmic transport, neuronal polarity, and even the secretion of tau, which is normally a cytosolic protein, to the extracellular space. In this review, we discuss tau misprocessing, toxicity and secretion in the context of normal tau functions in developing and mature neurons. We also compare tau cytopathology to that of other aggregation-prone proteins involved in neurodegeneration (alpha synuclein, prion protein, and APP). Finally, we consider potential mechanisms of intra- and interneuronal tau lesion spreading, an area of particular recent interest.
Collapse
Affiliation(s)
- Kerry L Gendreau
- Department of Biological Sciences, University of Massachusetts Lowell , Lowell, MA , USA
| | | |
Collapse
|
38
|
Using protein misfolding cyclic amplification generates a highly neurotoxic PrP dimer causing neurodegeneration. J Mol Neurosci 2013; 51:655-62. [PMID: 23771785 DOI: 10.1007/s12031-013-0039-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 05/27/2013] [Indexed: 10/26/2022]
Abstract
Under the "protein-only" hypothesis, prion-based diseases are proposed to result from an infectious agent that is an abnormal isoform of the prion protein in the scrapie form, PrP(Sc). However, since PrP(Sc) is highly insoluble and easily aggregates in vivo, this view appears to be overly simplistic, implying that the presence of PrP(Sc) may indirectly cause neurodegeneration through its intermediate soluble form. We generated a neurotoxic PrP dimer with partial pathogenic characteristics of PrP(Sc) by protein misfolding cyclic amplification in the presence of 1-palmitoyl-2-oleoylphosphatidylglycerol consisting of recombinant hamster PrP (23-231). After intracerebral injection of the PrP dimer, wild-type hamsters developed signs of neurodegeneration. Clinical symptoms, necropsy findings, and histopathological changes were very similar to those of transmissible spongiform encephalopathies. Additional investigation showed that the toxicity is primarily related to cellular apoptosis. All results suggested that we generated a new neurotoxic form of PrP, PrP dimer, which can cause neurodegeneration. Thus, our study introduces a useful model for investigating PrP-linked neurodegenerative mechanisms.
Collapse
|
39
|
Abstract
The human cellular prion protein (PrP(C)) is a glycosylphosphatidylinositol (GPI) anchored membrane glycoprotein with two N-glycosylation sites at residues 181 and 197. This protein migrates in several bands by Western blot analysis (WB). Interestingly, PNGase F treatment of human brain homogenates prior to the WB, which is known to remove the N-glycosylations, unexpectedly gives rise to two dominant bands, which are now known as C-terminal (C1) and N-terminal (N1) fragments. This resembles the β-amyloid precursor protein (APP) in Alzheimer disease (AD), which can be physiologically processed by α-, β-, and γ-secretases. The processing of APP has been extensively studied, while the identity of the cellular proteases involved in the proteolysis of PrP(C) and their possible role in prion biology has remained limited and controversial. Nevertheless, there is a strong correlation between the neurotoxicity caused by prion proteins and the blockade of their normal proteolysis. For example, expression of non-cleavable PrP(C) mutants in transgenic mice generates neurotoxicity, even in the absence of infectious prions, suggesting that PrP(C) proteolysis is physiologically and pathologically important. As many mouse models of prion diseases have recently been developed and the knowledge about the proteases responsible for the PrP(C) proteolysis is accumulating, we examine the historical experimental evidence and highlight recent studies that shed new light on this issue.
Collapse
|
40
|
Senatore A, Colleoni S, Verderio C, Restelli E, Morini R, Condliffe S, Bertani I, Mantovani S, Canovi M, Micotti E, Forloni G, Dolphin A, Matteoli M, Gobbi M, Chiesa R. Mutant PrP suppresses glutamatergic neurotransmission in cerebellar granule neurons by impairing membrane delivery of VGCC α(2)δ-1 Subunit. Neuron 2012; 74:300-13. [PMID: 22542184 PMCID: PMC3339322 DOI: 10.1016/j.neuron.2012.02.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2012] [Indexed: 01/17/2023]
Abstract
How mutant prion protein (PrP) leads to neurological dysfunction in genetic prion diseases is unknown. Tg(PG14) mice synthesize a misfolded mutant PrP which is partially retained in the neuronal endoplasmic reticulum (ER). As these mice age, they develop ataxia and massive degeneration of cerebellar granule neurons (CGNs). Here, we report that motor behavioral deficits in Tg(PG14) mice emerge before neurodegeneration and are associated with defective glutamate exocytosis from granule neurons due to impaired calcium dynamics. We found that mutant PrP interacts with the voltage-gated calcium channel α(2)δ-1 subunit, which promotes the anterograde trafficking of the channel. Owing to ER retention of mutant PrP, α(2)δ-1 accumulates intracellularly, impairing delivery of the channel complex to the cell surface. Thus, mutant PrP disrupts cerebellar glutamatergic neurotransmission by reducing the number of functional channels in CGNs. These results link intracellular PrP retention to synaptic dysfunction, indicating new modalities of neurotoxicity and potential therapeutic strategies.
Collapse
Affiliation(s)
- Assunta Senatore
- Dulbecco Telethon Institute, 20156 Milan, Italy, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Simona Colleoni
- Department of Biochemistry and Molecular Pharmacology, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Claudia Verderio
- Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche Institute of Neuroscience, University of Milan, 20129 Milan, Italy
| | - Elena Restelli
- Dulbecco Telethon Institute, 20156 Milan, Italy, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Raffaella Morini
- Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche Institute of Neuroscience, University of Milan, 20129 Milan, Italy
| | - Steven B. Condliffe
- Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche Institute of Neuroscience, University of Milan, 20129 Milan, Italy
| | - Ilaria Bertani
- Dulbecco Telethon Institute, 20156 Milan, Italy, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Susanna Mantovani
- Dulbecco Telethon Institute, 20156 Milan, Italy, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Mara Canovi
- Department of Biochemistry and Molecular Pharmacology, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Annette C. Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, WC1E6BT London, UK
| | - Michela Matteoli
- Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche Institute of Neuroscience, University of Milan, 20129 Milan, Italy
- Istituto Clinico Humanitas IRCCS, 20089 Milan, Italy
| | - Marco Gobbi
- Department of Biochemistry and Molecular Pharmacology, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Roberto Chiesa
- Dulbecco Telethon Institute, 20156 Milan, Italy, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Corresponding author
| |
Collapse
|
41
|
Margalith I, Suter C, Ballmer B, Schwarz P, Tiberi C, Sonati T, Falsig J, Nyström S, Hammarström P, Aslund A, Nilsson KPR, Yam A, Whitters E, Hornemann S, Aguzzi A. Polythiophenes inhibit prion propagation by stabilizing prion protein (PrP) aggregates. J Biol Chem 2012; 287:18872-87. [PMID: 22493452 DOI: 10.1074/jbc.m112.355958] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Luminescent conjugated polymers (LCPs) interact with ordered protein aggregates and sensitively detect amyloids of many different proteins, suggesting that they may possess antiprion properties. Here, we show that a variety of anionic, cationic, and zwitterionic LCPs reduced the infectivity of prion-containing brain homogenates and of prion-infected cerebellar organotypic cultured slices and decreased the amount of scrapie isoform of PrP(C) (PrP(Sc)) oligomers that could be captured in an avidity assay. Paradoxically, treatment enhanced the resistance of PrP(Sc) to proteolysis, triggered the compaction, and enhanced the resistance to proteolysis of recombinant mouse PrP(23-231) fibers. These results suggest that LCPs act as antiprion agents by transitioning PrP aggregates into structures with reduced frangibility. Moreover, ELISA on cerebellar organotypic cultured slices and in vitro conversion assays with mouse PrP(23-231) indicated that poly(thiophene-3-acetic acid) may additionally interfere with the generation of PrP(Sc) by stabilizing the conformation of PrP(C) or of a transition intermediate. Therefore, LCPs represent a novel class of antiprion agents whose mode of action appears to rely on hyperstabilization, rather than destabilization, of PrP(Sc) deposits.
Collapse
Affiliation(s)
- Ilan Margalith
- Institute of Neuropathology, University Hospital of Zurich, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Thackray AM, Muhammad F, Zhang C, Denyer M, Spiropoulos J, Crowther DC, Bujdoso R. Prion-induced toxicity in PrP transgenic Drosophila. Exp Mol Pathol 2012; 92:194-201. [PMID: 22314254 DOI: 10.1016/j.yexmp.2012.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 01/17/2012] [Indexed: 02/07/2023]
Abstract
Prion diseases are fatal transmissible neurodegenerative diseases of humans and various vertebrate species. In their natural hosts these conditions are characterised by prolonged incubation times prior to the onset of clinical signs of terminal disease. Accordingly, tractable models of mammalian prion disease are required in order to better understand the mechanisms of prion replication and prion-induced neurotoxicity. Transmission of prion diseases can occur across a species barrier and this is facilitated in recipients transgenic for the same PrP gene as the individual from which the infectious prions are derived. Here we have tested the hypothesis that exogenous ovine prions can induce neurotoxicity in Drosophila melanogaster transgenic for ovine PrP. Drosophila that expressed ovine PrP pan neuronally and inoculated with ovine prions at the larval stage by oral exposure to scrapie-infected sheep brain homogenate showed markedly accelerated locomotor and survival defects. ARQ PrP transgenic Drosophila exposed to scrapie-infected brain homogenate showed a significant and progressive reduction in locomotor activity compared to similar flies exposed to normal sheep brain homogenate. The prion-induced locomotor defect was accompanied by the accumulation of potentially misfolded PrP in the brains of prion-inoculated flies. VRQ PrP transgenic Drosophila, which expressed less ovine PrP than ARQ flies, showed a reduced median survival compared to similar flies exposed to normal sheep brain homogenate. These prion-induced phenotypic effects were PrP-mediated since ovine prions were not toxic in non-PrP transgenic control flies. Our observations provide the basis of an invertebrate model of transmissible mammalian prion disease.
Collapse
Affiliation(s)
- Alana M Thackray
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, Cambridge, CB3 OES, UK
| | | | | | | | | | | | | |
Collapse
|
43
|
Faucheux BA, Morain E, Diouron V, Brandel JP, Salomon D, Sazdovitch V, Privat N, Laplanche JL, Hauw JJ, Haïk S. Quantification of surviving cerebellar granule neurones and abnormal prion protein (PrPSc) deposition in sporadic Creutzfeldt-Jakob disease supports a pathogenic role for small PrPSc deposits common to the various molecular subtypes. Neuropathol Appl Neurobiol 2011; 37:500-12. [PMID: 21450052 DOI: 10.1111/j.1365-2990.2011.01179.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AIMS Neuronal death is a major neuropathological hallmark in prion diseases. The association between the accumulation of the disease-related prion protein (PrP(Sc) ) and neuronal loss varies within the wide spectrum of prion diseases and their experimental models. In this study, we investigated the relationships between neuronal loss and PrP(Sc) deposition in the cerebellum from cases of the six subtypes of sporadic Creutzfeldt-Jakob disease (sCJD; n=100) that can be determined according to the M129V polymorphism of the human prion protein gene (PRNP) and PrP(Sc) molecular types. METHODS The numerical density of neurones was estimated with a computer-assisted image analysis system and the accumulation of PrP(Sc) deposits was scored. RESULTS The scores of PrP(Sc) immunoreactive deposits of the punctate type (synaptic type) were correlated with neurone counts - the higher the score the higher the neuronal loss - in all sCJD subtypes. Large 5- to 50-µm-wide deposits (focal type) were found in sCJD-MV2 and sCJD-VV2 subtypes, and occasionally in a few cases of the other studied groups. By contrast, the highest scores for 5- to 50-µm-wide deposits observed in sCJD-MV2 subtype were not associated with higher neuronal loss. In addition, these scores were inversely correlated with neuronal counts in the sCJD-VV2 subtype. CONCLUSIONS These results support a putative pathogenic role for small PrP(Sc) deposits common to the various sCJD subtypes. Furthermore, the observation of a lower loss of neurones associated with PrP(Sc) type-2 large deposits is consistent with a possible 'protective' role of aggregated deposits in both sCJD-MV2 and sCJD-VV2 subtypes.
Collapse
Affiliation(s)
- B A Faucheux
- Assistance Publique - Hôpitaux de Paris (AP-HP), Laboratoire de Neuropathologie, Hôpital de la Salpêtrière, 47 boulevard de l'Hôpital Centre de Recherche de l'Institut du Cerveau et de la Moelle, INSERM UMRS975, CNRS UMR7225, Paris, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Li B, Qing L, Yan J, Kong Q. Instability of the octarepeat region of the human prion protein gene. PLoS One 2011; 6:e26635. [PMID: 22028931 PMCID: PMC3197570 DOI: 10.1371/journal.pone.0026635] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 09/30/2011] [Indexed: 01/24/2023] Open
Abstract
Prion diseases are a family of unique fatal transmissible neurodegenerative diseases that affect humans and many animals. Sporadic Creutzfeldt-Jakob disease (sCJD) is the most common prion disease in humans, accounting for 85–90% of all human prion cases, and exhibits a high degree of diversity in phenotypes. The etiology of sCJD remains to be elucidated. The human prion protein gene has an octapeptide repeat region (octarepeats) that normally contains 5 repeats of 24–27 bp (1 nonapeptide and 4 octapeptide coding sequences). An increase of the octarepeat numbers to six or more or a decrease of the octarepeat number to three is linked to genetic prion diseases with heterogeneous phenotypes in humans. Here we report that the human octarepeat region is prone to either contraction or expansion when subjected to PCR amplification in vitro using Taq or Pwo polymerase and when replicated in wild type E. coli cells. Octarepeat insertion mutants were even less stable, and the mutation rate for the wild type octarepeats was much higher when replicated in DNA mismatch repair-deficient E.coli cells. All observed octarepeat mutants resulting from DNA replication in E.coli were contained in head-to-head plasmid dimers and DNA mfold analysis (http://mfold.rna.albany.edu/?q=mfold/DNA-Folding-Form) indicates that both DNA strands of the octarepeat region would likely form multiple stable hairpin structures, suggesting that the octarepeat sequence may form stable hairpin structures during DNA replication or repair to cause octarepeat instability. These results provide the first evidence supporting a somatic octarepeat mutation-based model for human sCJD etiology: 1) the instability of the octarepeat region leads to accumulation of somatic octarepeat mutations in brain cells during development and aging, 2) this instability is augmented by compromised DNA mismatch repair in aged cells, and 3) eventually some of the octarepeat mutation-containing brain cells start spontaneous de novo prion formation and replication to initiate sCJD.
Collapse
Affiliation(s)
- Baiya Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | | | | | | |
Collapse
|
45
|
Jeffrey M, Scholes SFE, Martin S, McGovern G, Sisó S, González L. Increased immunohistochemical labelling for prion protein occurs in diverse neurological disorders of sheep: relevance for normal cellular PrP function. J Comp Pathol 2011; 147:46-54. [PMID: 22000036 DOI: 10.1016/j.jcpa.2011.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 08/12/2011] [Accepted: 08/19/2011] [Indexed: 11/17/2022]
Abstract
The classical prion diseases (e.g. scrapie of sheep and goats and bovine spongiform encephalopathy of cattle) are characterized by the accumulation of abnormal forms of the prion protein (PrP), usually recognized by their relative resistance to proteolysis compared with the physiological cellular forms of PrP. However, novel prion diseases have been detected in sheep, cattle and man, in which the abnormal PrP has less resistance to proteolysis than identified previously. These more subtle differences between abnormal and normal forms of PrP can be problematic in routine diagnostic tests and raise questions in respect of the range of PrP disorders. Abnormal accumulations of PrP in atypical and classical prion diseases can be recognized by immunohistochemistry. To determine whether altered PrP expression or trafficking might occur in nosological entities not previously connected with prion disease, the brains of sheep affected with diverse neurological conditions were examined for evidence of altered PrP labelling. Such altered immunolabelling was detected in association with either basic lesions or specific diseases. Some reactive glial cells and degenerate neurons found in several different recognized disorders and non-specific inflammatory processes were associated with abnormal PrP labelling, which was absent from brains of healthy, age-matched sheep. The results agree with previous indications that normal PrP function may be linked with the oxidative stress response, but the data also suggest that PrP functions are more extensive than simple protective responses against stress insults.
Collapse
Affiliation(s)
- M Jeffrey
- Animal Health and Veterinary Laboratories Agency, Lasswade Veterinary Laboratory, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 OPZ, Scotland, UK.
| | | | | | | | | | | |
Collapse
|
46
|
Jeffrey M, McGovern G, Chambers EV, King D, González L, Manson JC, Ghetti B, Piccardo P, Barron RM. Mechanism of PrP-amyloid formation in mice without transmissible spongiform encephalopathy. Brain Pathol 2011; 22:58-66. [PMID: 21645162 DOI: 10.1111/j.1750-3639.2011.00508.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gerstmann-Sträussler-Scheinker (GSS) P102L disease is a familial form of a transmissible spongiform encephalopathy (TSE) that can present with or without vacuolation of neuropil. Inefficient disease transmission into 101LL transgenic mice was previously observed from GSS P102L without vacuolation. However, several aged, healthy mice had large plaques composed of abnormal prion protein (PrP(d)). Here we perform the ultrastructural characterization of such plaques and compare them with PrP(d) aggregates found in TSE caused by an infectious mechanism. PrP(d) plaques in 101LL mice varied in maturity, with some being composed of deposits without visible amyloid fibrils. PrP(d) was present on cell membranes in the vicinity of all types of plaques. In contrast to the unicentric plaques seen in infectious murine scrapie, the plaques seen in the current model were multicentric and were initiated by protofibrillar forms of PrP(d) situated on oligodendroglia, astrocytes and neuritic cell membranes. We speculate that the initial conversion process leading to plaque formation begins with membrane-bound PrP(C) but that subsequent fibrillization does not require membrane attachment. We also observed that the membrane alterations consistently seen in murine scrapie and other infectious TSEs were not present in 101LL mice with plaques, suggesting differences in the pathogenesis of these conditions.
Collapse
Affiliation(s)
- Martin Jeffrey
- Animal Health Veterinary Laboratories Agency, Pentlands Science Park, Bush Loan Penicuik, Midlothian, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Misfolded PrP impairs the UPS by interaction with the 20S proteasome and inhibition of substrate entry. EMBO J 2011; 30:3065-77. [PMID: 21743439 DOI: 10.1038/emboj.2011.224] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 06/16/2011] [Indexed: 01/07/2023] Open
Abstract
Prion diseases are associated with the conversion of cellular prion protein (PrP(C)) to toxic β-sheet isoforms (PrP(Sc)), which are reported to inhibit the ubiquitin-proteasome system (UPS). Accordingly, UPS substrates accumulate in prion-infected mouse brains, suggesting impairment of the 26S proteasome. A direct interaction between its 20S core particle and PrP isoforms was demonstrated by immunoprecipitation. β-PrP aggregates associated with the 20S particle, but did not impede binding of the PA26 complex, suggesting that the aggregates do not bind to its ends. Aggregated β-PrP reduced the 20S proteasome's basal peptidase activity, and the enhanced activity induced by C-terminal peptides from the 19S ATPases or by the 19S regulator itself, including when stimulated by polyubiquitin conjugates. However, the 20S proteasome was not inhibited when the gate in the α-ring was open due to a truncation mutation or by association with PA26/PA28. These PrP aggregates inhibit by stabilising the closed conformation of the substrate entry channel. A similar inhibition of substrate entry into the proteasome may occur in other neurodegenerative diseases where misfolded β-sheet-rich proteins accumulate.
Collapse
|
48
|
Quaglio E, Restelli E, Garofoli A, Dossena S, De Luigi A, Tagliavacca L, Imperiale D, Migheli A, Salmona M, Sitia R, Forloni G, Chiesa R. Expression of mutant or cytosolic PrP in transgenic mice and cells is not associated with endoplasmic reticulum stress or proteasome dysfunction. PLoS One 2011; 6:e19339. [PMID: 21559407 PMCID: PMC3084828 DOI: 10.1371/journal.pone.0019339] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 03/27/2011] [Indexed: 12/20/2022] Open
Abstract
The cellular pathways activated by mutant prion protein (PrP) in genetic prion diseases, ultimately leading to neuronal dysfunction and degeneration, are not known. Several mutant PrPs misfold in the early secretory pathway and reside longer in the endoplasmic reticulum (ER) possibly stimulating ER stress-related pathogenic mechanisms. To investigate whether mutant PrP induced maladaptive responses, we checked key elements of the unfolded protein response (UPR) in transgenic mice, primary neurons and transfected cells expressing two different mutant PrPs. Because ER stress favors the formation of untranslocated PrP that might aggregate in the cytosol and impair proteasome function, we also measured the activity of the ubiquitin proteasome system (UPS). Molecular, biochemical and immunohistochemical analyses found no increase in the expression of UPR-regulated genes, such as Grp78/Bip, CHOP/GADD153, or ER stress-dependent splicing of the mRNA encoding the X-box-binding protein 1. No alterations in UPS activity were detected in mutant mouse brains and primary neurons using the UbG76V-GFP reporter and a new fluorogenic peptide for monitoring proteasomal proteolytic activity in vivo. Finally, there was no loss of proteasome function in neurons in which endogenous PrP was forced to accumulate in the cytosol by inhibiting cotranslational translocation. These results indicate that neither ER stress, nor perturbation of proteasome activity plays a major pathogenic role in prion diseases.
Collapse
Affiliation(s)
- Elena Quaglio
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Elena Restelli
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Anna Garofoli
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Sara Dossena
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Ada De Luigi
- Department of Biochemistry and Molecular Pharmacology, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Luigina Tagliavacca
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Università Vita-Salute San Raffaele, Milan, Italy
| | - Daniele Imperiale
- Neurology Unit, Human Prion Diseases Center D.O.M.P., Maria Vittoria Hospital, Torino, Italy
| | - Antonio Migheli
- Neurology Unit, Human Prion Diseases Center D.O.M.P., Maria Vittoria Hospital, Torino, Italy
| | - Mario Salmona
- Department of Biochemistry and Molecular Pharmacology, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Roberto Sitia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Università Vita-Salute San Raffaele, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Roberto Chiesa
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
- * E-mail:
| |
Collapse
|
49
|
Wadsworth JDF, Asante EA, Collinge J. Review: contribution of transgenic models to understanding human prion disease. Neuropathol Appl Neurobiol 2011; 36:576-97. [PMID: 20880036 PMCID: PMC3017745 DOI: 10.1111/j.1365-2990.2010.01129.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Transgenic mice expressing human prion protein in the absence of endogenous mouse prion protein faithfully replicate human prions. These models reproduce all of the key features of human disease, including long clinically silent incubation periods prior to fatal neurodegeneration with neuropathological phenotypes that mirror human prion strain diversity. Critical contributions to our understanding of human prion disease pathogenesis and aetiology have only been possible through the use of transgenic mice. These models have provided the basis for the conformational selection model of prion transmission barriers and have causally linked bovine spongiform encephalopathy with variant Creutzfeldt-Jakob disease. In the future these models will be essential for evaluating newly identified potentially zoonotic prion strains, for validating effective methods of prion decontamination and for developing effective therapeutic treatments for human prion disease.
Collapse
Affiliation(s)
- J D F Wadsworth
- MRC Prion Unit and Department of Neurodegenerative Disease, Institute of Neurology, University College London, National Hospital for Neurology and Neurosurgery, London, UK.
| | | | | |
Collapse
|
50
|
Abstract
Prion diseases in humans and animals are characterized by progressive neurodegeneration and the formation of infectious particles called prions. Both features are intimately linked to a conformational transition of the cellular prion protein (PrP(C)) into aberrantly folded conformers with neurotoxic and self-replicating activities. Interestingly, there is increasing evidence that the infectious and neurotoxic properties of PrP conformers are not necessarily coupled. Transgenic mouse models revealed that some PrP mutants interfere with neuronal function in the absence of infectious prions. Vice versa, propagation of prions can occur without causing neurotoxicity. Consequently, it appears plausible that two partially independent pathways exist, one pathway leading to the propagation of infectious prions and another one that mediates neurotoxic signaling. In this review we will summarize current knowledge of neurotoxic PrP conformers and discuss the role of PrP(C) as a mediator of both stress-protective and neurotoxic signaling cascades.
Collapse
|