1
|
Eid S, Lee S, Verkuyl CE, Almanza D, Hanna J, Shenouda S, Belotserkovsky A, Zhao W, Watts JC. The importance of prion research. Biochem Cell Biol 2024. [PMID: 38996387 DOI: 10.1139/bcb-2024-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024] Open
Abstract
Over the past four decades, prion diseases have received considerable research attention owing to their potential to be transmitted within and across species as well as their consequences for human and animal health. The unprecedented nature of prions has led to the discovery of a paradigm of templated protein misfolding that underlies a diverse range of both disease-related and normal biological processes. Indeed, the "prion-like" misfolding and propagation of protein aggregates is now recognized as a common underlying disease mechanism in human neurodegenerative disorders such as Alzheimer's and Parkinson's disease, and the prion principle has led to the development of novel diagnostic and therapeutic strategies for these illnesses. Despite these advances, research into the fundamental biology of prion diseases has declined, likely due to their rarity and the absence of an acute human health crisis. Given the past translational influence, continued research on the etiology, pathogenesis, and transmission of prion disease should remain a priority. In this review, we highlight several important "unsolved mysteries" in the prion disease research field and how solving them may be crucial for the development of effective therapeutics, preventing future outbreaks of prion disease, and understanding the pathobiology of more common human neurodegenerative disorders.
Collapse
Affiliation(s)
- Shehab Eid
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Seojin Lee
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Claire E Verkuyl
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Dustin Almanza
- Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Joseph Hanna
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Sandra Shenouda
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Ari Belotserkovsky
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Wenda Zhao
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
Zayed M, Kook SH, Jeong BH. Potential Therapeutic Use of Stem Cells for Prion Diseases. Cells 2023; 12:2413. [PMID: 37830627 PMCID: PMC10571911 DOI: 10.3390/cells12192413] [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: 08/30/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023] Open
Abstract
Prion diseases are neurodegenerative disorders that are progressive, incurable, and deadly. The prion consists of PrPSc, the misfolded pathogenic isoform of the cellular prion protein (PrPC). PrPC is involved in a variety of physiological functions, including cellular proliferation, adhesion, differentiation, and neural development. Prion protein is expressed on the membrane surface of a variety of stem cells (SCs), where it plays an important role in the pluripotency and self-renewal matrix, as well as in SC differentiation. SCs have been found to multiply the pathogenic form of the prion protein, implying their potential as an in vitro model for prion diseases. Furthermore, due to their capability to self-renew, differentiate, immunomodulate, and regenerate tissue, SCs are prospective cell treatments in many neurodegenerative conditions, including prion diseases. Regenerative medicine has become a new revolution in disease treatment in recent years, particularly with the introduction of SC therapy. Here, we review the data demonstrating prion diseases' biology and molecular mechanism. SC biology, therapeutic potential, and its role in understanding prion disease mechanisms are highlighted. Moreover, we summarize preclinical studies that use SCs in prion diseases.
Collapse
Affiliation(s)
- Mohammed Zayed
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea;
- Department of Bioactive Material Sciences, Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Department of Surgery, College of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - Sung-Ho Kook
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea;
- Department of Bioactive Material Sciences, Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|
3
|
Walters RO, Haigh CL. Organoids for modeling prion diseases. Cell Tissue Res 2023; 392:97-111. [PMID: 35088182 PMCID: PMC9329493 DOI: 10.1007/s00441-022-03589-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/19/2022] [Indexed: 11/25/2022]
Abstract
Human cerebral organoids are an exciting and novel model system emerging in the field of neurobiology. Cerebral organoids are spheres of self-organizing, neuronal lineage tissue that can be differentiated from human pluripotent stem cells and that present the possibility of on-demand human neuronal cultures that can be used for non-invasively investigating diseases affecting the brain. Compared with existing humanized cell models, they provide a more comprehensive replication of the human cerebral environment. The potential of the human cerebral organoid model is only just beginning to be elucidated, but initial studies have indicated that they could prove to be a valuable model for neurodegenerative diseases such as prion disease. The application of the cerebral organoid model to prion disease, what has been learned so far and the future potential of this model are discussed in this review.
Collapse
Affiliation(s)
- Ryan O Walters
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - Cathryn L Haigh
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA.
| |
Collapse
|
4
|
Arshad H, Watts JC. Genetically engineered cellular models of prion propagation. Cell Tissue Res 2022; 392:63-80. [PMID: 35581386 DOI: 10.1007/s00441-022-03630-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/26/2022] [Indexed: 11/02/2022]
Abstract
For over three decades, cultured cells have been a useful tool for dissecting the molecular details of prion replication and the identification of candidate therapeutics for prion disease. A major issue limiting the translatability of these studies has been the inability to reliably propagate disease-relevant, non-mouse strains of prions in cells relevant to prion pathogenesis. In recent years, fueled by advances in gene editing technology, it has become possible to propagate prions from hamsters, cervids, and sheep in immortalized cell lines originating from the central nervous system. In particular, the use of CRISPR-Cas9-mediated gene editing to generate versions of prion-permissive cell lines that lack endogenous PrP expression has provided a blank canvas upon which re-expression of PrP leads to species-matched susceptibility to prion infection. When coupled with the ability to propagate prions in cells or organoids derived from stem cells, these next-generation cellular models should provide an ideal paradigm for identifying small molecules and other biological therapeutics capable of interfering with prion replication in animal and human prion disorders. In this review, we summarize recent advances that have widened the spectrum of prion strains that can be propagated in cultured cells and cutting-edge tissue-based models.
Collapse
Affiliation(s)
- Hamza Arshad
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower Rm. 4KD481, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada.,Department of Biochemistry, University of Toronto, 1 King's College Circle, Medical Sciences Building Rm. 5207, Toronto, ON, M5S 1A8, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower Rm. 4KD481, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada. .,Department of Biochemistry, University of Toronto, 1 King's College Circle, Medical Sciences Building Rm. 5207, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
5
|
Khadka A, Spiers JG, Cheng L, Hill AF. Extracellular vesicles with diagnostic and therapeutic potential for prion diseases. Cell Tissue Res 2022; 392:247-267. [PMID: 35394216 PMCID: PMC10113352 DOI: 10.1007/s00441-022-03621-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/25/2022] [Indexed: 12/14/2022]
Abstract
Prion diseases (PrD) or transmissible spongiform encephalopathies (TSE) are invariably fatal and pathogenic neurodegenerative disorders caused by the self-propagated misfolding of cellular prion protein (PrPC) to the neurotoxic pathogenic form (PrPTSE) via a yet undefined but profoundly complex mechanism. Despite several decades of research on PrD, the basic understanding of where and how PrPC is transformed to the misfolded, aggregation-prone and pathogenic PrPTSE remains elusive. The primary clinical hallmarks of PrD include vacuolation-associated spongiform changes and PrPTSE accumulation in neural tissue together with astrogliosis. The difficulty in unravelling the disease mechanisms has been related to the rare occurrence and long incubation period (over decades) followed by a very short clinical phase (few months). Additional challenge in unravelling the disease is implicated to the unique nature of the agent, its complexity and strain diversity, resulting in the heterogeneity of the clinical manifestations and potentially diverse disease mechanisms. Recent advances in tissue isolation and processing techniques have identified novel means of intercellular communication through extracellular vesicles (EVs) that contribute to PrPTSE transmission in PrD. This review will comprehensively discuss PrPTSE transmission and neurotoxicity, focusing on the role of EVs in disease progression, biomarker discovery and potential therapeutic agents for the treatment of PrD.
Collapse
Affiliation(s)
- Arun Khadka
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Jereme G Spiers
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Lesley Cheng
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Andrew F Hill
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia. .,Institute for Health and Sport, Victoria University, Footscray, VIC, Australia.
| |
Collapse
|
6
|
Gene-Edited Cell Models to Study Chronic Wasting Disease. Viruses 2022; 14:v14030609. [PMID: 35337016 PMCID: PMC8950194 DOI: 10.3390/v14030609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 11/17/2022] Open
Abstract
Prion diseases are fatal infectious neurodegenerative disorders affecting both humans and animals. They are caused by the misfolded isoform of the cellular prion protein (PrPC), PrPSc, and currently no options exist to prevent or cure prion diseases. Chronic wasting disease (CWD) in deer, elk and other cervids is considered the most contagious prion disease, with extensive shedding of infectivity into the environment. Cell culture models provide a versatile platform for convenient quantification of prions, for studying the molecular and cellular biology of prions, and for performing high-throughput screening of potential therapeutic compounds. Unfortunately, only a very limited number of cell lines are available that facilitate robust and persistent propagation of CWD prions. Gene-editing using programmable nucleases (e.g., CRISPR-Cas9 (CC9)) has proven to be a valuable tool for high precision site-specific gene modification, including gene deletion, insertion, and replacement. CC9-based gene editing was used recently for replacing the PrP gene in mouse and cell culture models, as efficient prion propagation usually requires matching sequence homology between infecting prions and prion protein in the recipient host. As expected, such gene-editing proved to be useful for developing CWD models. Several transgenic mouse models were available that propagate CWD prions effectively, however, mostly fail to reproduce CWD pathogenesis as found in the cervid host, including CWD prion shedding. This is different for the few currently available knock-in mouse models that seem to do so. In this review, we discuss the available in vitro and in vivo models of CWD, and the impact of gene-editing strategies.
Collapse
|
7
|
Heumüller SE, Hornberger AC, Hebestreit AS, Hossinger A, Vorberg IM. Propagation and Dissemination Strategies of Transmissible Spongiform Encephalopathy Agents in Mammalian Cells. Int J Mol Sci 2022; 23:ijms23062909. [PMID: 35328330 PMCID: PMC8949484 DOI: 10.3390/ijms23062909] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 01/08/2023] Open
Abstract
Transmissible spongiform encephalopathies or prion disorders are fatal infectious diseases that cause characteristic spongiform degeneration in the central nervous system. The causative agent, the so-called prion, is an unconventional infectious agent that propagates by converting the host-encoded cellular prion protein PrP into ordered protein aggregates with infectious properties. Prions are devoid of coding nucleic acid and thus rely on the host cell machinery for propagation. While it is now established that, in addition to PrP, other cellular factors or processes determine the susceptibility of cell lines to prion infection, exact factors and cellular processes remain broadly obscure. Still, cellular models have uncovered important aspects of prion propagation and revealed intercellular dissemination strategies shared with other intracellular pathogens. Here, we summarize what we learned about the processes of prion invasion, intracellular replication and subsequent dissemination from ex vivo cell models.
Collapse
Affiliation(s)
- Stefanie-Elisabeth Heumüller
- Laboratory of Prion Cell Biology, German Center for Neurodegenerative Diseases Bonn (DZNE e.V.), Venusberg-Campus 1/99, 53127 Bonn, Germany; (S.-E.H.); (A.C.H.); (A.S.H.); (A.H.)
| | - Annika C. Hornberger
- Laboratory of Prion Cell Biology, German Center for Neurodegenerative Diseases Bonn (DZNE e.V.), Venusberg-Campus 1/99, 53127 Bonn, Germany; (S.-E.H.); (A.C.H.); (A.S.H.); (A.H.)
| | - Alina S. Hebestreit
- Laboratory of Prion Cell Biology, German Center for Neurodegenerative Diseases Bonn (DZNE e.V.), Venusberg-Campus 1/99, 53127 Bonn, Germany; (S.-E.H.); (A.C.H.); (A.S.H.); (A.H.)
| | - André Hossinger
- Laboratory of Prion Cell Biology, German Center for Neurodegenerative Diseases Bonn (DZNE e.V.), Venusberg-Campus 1/99, 53127 Bonn, Germany; (S.-E.H.); (A.C.H.); (A.S.H.); (A.H.)
| | - Ina M. Vorberg
- Laboratory of Prion Cell Biology, German Center for Neurodegenerative Diseases Bonn (DZNE e.V.), Venusberg-Campus 1/99, 53127 Bonn, Germany; (S.-E.H.); (A.C.H.); (A.S.H.); (A.H.)
- German Center for Neurodegenerative Diseases (DZNE), Rheinische Friedrich-Wilhelms-Universität Bonn, Siegmund-Freud-Str. 25, 53127 Bonn, Germany
- Correspondence:
| |
Collapse
|
8
|
Pineau H, Sim VL. From Cell Culture to Organoids-Model Systems for Investigating Prion Strain Characteristics. Biomolecules 2021; 11:biom11010106. [PMID: 33466947 PMCID: PMC7830147 DOI: 10.3390/biom11010106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Prion diseases are the hallmark protein folding neurodegenerative disease. Their transmissible nature has allowed for the development of many different cellular models of disease where prion propagation and sometimes pathology can be induced. This review examines the range of simple cell cultures to more complex neurospheres, organoid, and organotypic slice cultures that have been used to study prion disease pathogenesis and to test therapeutics. We highlight the advantages and disadvantages of each system, giving special consideration to the importance of strains when choosing a model and when interpreting results, as not all systems propagate all strains, and in some cases, the technique used, or treatment applied, can alter the very strain properties being studied.
Collapse
Affiliation(s)
- Hailey Pineau
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada;
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Valerie L. Sim
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada;
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Correspondence:
| |
Collapse
|
9
|
Arshad H, Bourkas MEC, Watts JC. The utility of bank voles for studying prion disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:179-211. [PMID: 32958232 DOI: 10.1016/bs.pmbts.2020.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The transmission of prions between species is typically an inefficient process due to the species barrier, which represents incompatibility between prion seed and substrate molecules. Bank voles (Myodes glareolus) are an exception to this rule, as they are susceptible to a diverse range of prion strains from many different animal species. In particular, bank voles can be efficiently infected with most types of human prions and have played a critical role in validating variably protease-sensitive prionopathy (VPSPr) and certain forms of Gerstmann-Sträussler-Scheinker (GSS) disease as bona fide prion disorders rather than non-transmissible proteinopathies. The bank vole prion protein (BVPrP) confers a "universal prion acceptor" phenotype when expressed in mice and when used as a substrate for in vitro prion amplification assays, indicating that the unique prion transmission properties of bank voles are mediated by BVPrP. Over-expression of BVPrP in mice can also promote the spontaneous development of prion disease, indicating that BVPrP is intrinsically prone to both spontaneous and template-directed misfolding. Here, we discuss the utility of bank voles and BVPrP for prion research and how they have provided new tools for establishing rapid animal bioassays, modeling spontaneous prion disease, standardizing prion diagnostics, and understanding the molecular basis of the species barrier.
Collapse
Affiliation(s)
- Hamza Arshad
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Matthew E C Bourkas
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
10
|
Krance SH, Luke R, Shenouda M, Israwi AR, Colpitts SJ, Darwish L, Strauss M, Watts JC. Cellular models for discovering prion disease therapeutics: Progress and challenges. J Neurochem 2020; 153:150-172. [PMID: 31943194 DOI: 10.1111/jnc.14956] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Abstract
Prions, which cause fatal neurodegenerative disorders such as Creutzfeldt-Jakob disease, are misfolded and infectious protein aggregates. Currently, there are no treatments available to halt or even delay the progression of prion disease in the brain. The infectious nature of prions has resulted in animal paradigms that accurately recapitulate all aspects of prion disease, and these have proven to be instrumental for testing the efficacy of candidate therapeutics. Nonetheless, infection of cultured cells with prions provides a much more powerful system for identifying molecules capable of interfering with prion propagation. Certain lines of cultured cells can be chronically infected with various types of mouse prions, and these models have been used to unearth candidate anti-prion drugs that are at least partially efficacious when administered to prion-infected rodents. However, these studies have also revealed that not all types of prions are equal, and that drugs active against mouse prions are not necessarily effective against prions from other species. Despite some recent progress, the number of cellular models available for studying non-mouse prions remains limited. In particular, human prions have proven to be particularly challenging to propagate in cultured cells, which has severely hindered the discovery of drugs for Creutzfeldt-Jakob disease. In this review, we summarize the cellular models that are presently available for discovering and testing drugs capable of blocking the propagation of prions and highlight challenges that remain on the path towards developing therapies for prion disease.
Collapse
Affiliation(s)
- Saffire H Krance
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Russell Luke
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Marc Shenouda
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Ahmad R Israwi
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Sarah J Colpitts
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Lina Darwish
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Maximilian Strauss
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Ellett LJ, Revill ZT, Koo YQ, Lawson VA. Strain variation in treatment and prevention of human prion diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:121-145. [PMID: 32958230 DOI: 10.1016/bs.pmbts.2020.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
Transmissible spongiform encephalopathies or prion diseases describe a number of different human disorders that differ in their clinical phenotypes, which are nonetheless united by their transmissible nature and common pathology. Clinical variation in the absence of a conventional infectious agent is believed to be encoded by different conformations of the misfolded prion protein. This misfolded protein is the target of methods designed to prevent disease transmission in a surgical setting and reduction of the misfolded seed or preventing its continued propagation have been the focus of therapeutic strategies. It is therefore possible that strain variation may influence the efficacy of prevention and treatment approaches. Historically, an understanding of prion disease transmission and pathogenesis has been focused on research tools developed using agriculturally relevant strains of prion disease. However, an increased understanding of the molecular biology of human prion disorders has highlighted differences not only between different forms of the disease affecting humans and animals but also within diseases such as Creutzfeldt-Jakob Disease (CJD), which is represented by several sporadic CJD specific conformations and an additional conformation associated with variant CJD. In this chapter we will discuss whether prion strain variation can affect the efficacy of methods used to decontaminate prions and whether strain variation in pre-clinical models of prion disease can be used to identify therapeutic strategies that have the best possible chance of success in the clinic.
Collapse
Affiliation(s)
- Laura J Ellett
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, Australia
| | - Zoe T Revill
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, Australia
| | - Yong Qian Koo
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, Australia
| | - Victoria A Lawson
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
12
|
Mays CE, Armijo E, Morales R, Kramm C, Flores A, Tiwari A, Bian J, Telling GC, Pandita TK, Hunt CR, Soto C. Prion disease is accelerated in mice lacking stress-induced heat shock protein 70 (HSP70). J Biol Chem 2019; 294:13619-13628. [PMID: 31320473 DOI: 10.1074/jbc.ra118.006186] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 06/28/2019] [Indexed: 01/09/2023] Open
Abstract
Prion diseases are a group of incurable neurodegenerative disorders that affect humans and animals via infection with proteinaceous particles called prions. Prions are composed of PrPSc, a misfolded version of the cellular prion protein (PrPC). During disease progression, PrPSc replicates by interacting with PrPC and inducing its conversion to PrPSc As PrPSc accumulates, cellular stress mechanisms are activated to maintain cellular proteostasis, including increased protein chaperone levels. However, the exact roles of several of these chaperones remain unclear. Here, using various methodologies to monitor prion replication (i.e. protein misfolding cyclic amplification and cellular and animal infectivity bioassays), we studied the potential role of the molecular chaperone heat shock protein 70 (HSP70) in prion replication in vitro and in vivo Our results indicated that pharmacological induction of the heat shock response in cells chronically infected with prions significantly decreased PrPSc accumulation. We also found that HSP70 alters prion replication in vitro More importantly, prion infection of mice lacking the genes encoding stress-induced HSP70 exhibited accelerated prion disease progression compared with WT mice. In parallel with HSP70 being known to respond to endogenous and exogenous stressors such as heat, infection, toxicants, and ischemia, our results indicate that HSP70 may also play an important role in suppressing or delaying prion disease progression, opening opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Charles E Mays
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School, Houston, Texas 77030
| | - Enrique Armijo
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School, Houston, Texas 77030.,Facultad de Medicina, Universidad de los Andes, Av. San Carlos de Apoquindo, 2200 Las Condes, Santiago, Chile
| | - Rodrigo Morales
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School, Houston, Texas 77030
| | - Carlos Kramm
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School, Houston, Texas 77030.,Facultad de Medicina, Universidad de los Andes, Av. San Carlos de Apoquindo, 2200 Las Condes, Santiago, Chile
| | - Andrea Flores
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School, Houston, Texas 77030
| | - Anjana Tiwari
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030
| | - Jifeng Bian
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523
| | - Glenn C Telling
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523
| | - Tej K Pandita
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030
| | - Clayton R Hunt
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030
| | - Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School, Houston, Texas 77030 .,Facultad de Medicina, Universidad de los Andes, Av. San Carlos de Apoquindo, 2200 Las Condes, Santiago, Chile
| |
Collapse
|
13
|
Foliaki ST, Lewis V, Islam AMT, Ellett LJ, Senesi M, Finkelstein DI, Roberts B, Lawson VA, Adlard PA, Collins SJ. Early existence and biochemical evolution characterise acutely synaptotoxic PrPSc. PLoS Pathog 2019; 15:e1007712. [PMID: 30970042 PMCID: PMC6490942 DOI: 10.1371/journal.ppat.1007712] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 04/30/2019] [Accepted: 03/18/2019] [Indexed: 11/19/2022] Open
Abstract
Although considerable evidence supports that misfolded prion protein (PrPSc) is the principal component of “prions”, underpinning both transmissibility and neurotoxicity, clear consensus around a number of fundamental aspects of pathogenesis has not been achieved, including the time of appearance of neurotoxic species during disease evolution. Utilizing a recently reported electrophysiology paradigm, we assessed the acute synaptotoxicity of ex vivo PrPSc prepared as crude homogenates from brains of M1000 infected wild-type mice (cM1000) harvested at time-points representing 30%, 50%, 70% and 100% of the terminal stage of disease (TSD). Acute synaptotoxicity was assessed by measuring the capacity of cM1000 to impair hippocampal CA1 region long-term potentiation (LTP) and post-tetanic potentiation (PTP) in explant slices. Of particular note, cM1000 from 30% of the TSD was able to cause significant impairment of LTP and PTP, with the induced failure of LTP increasing over subsequent time-points while the capacity of cM1000 to induce PTP failure appeared maximal even at this early stage of disease progression. Evidence that the synaptotoxicity directly related to PrP species was demonstrated by the significant rescue of LTP dysfunction at each time-point through immuno-depletion of >50% of total PrP species from cM1000 preparations. Moreover, similar to our previous observations at the terminal stage of M1000 prion disease, size fractionation chromatography revealed that capacity for acute synpatotoxicity correlated with predominance of oligomeric PrP species in infected brains across all time points, with the profile appearing maximised by 50% of the TSD. Using enhanced sensitivity western blotting, modestly proteinase K (PK)-resistant PrPSc was detectable at very low levels in cM1000 at 30% of the TSD, becoming robustly detectable by 70% of the TSD at which time substantial levels of highly PK-resistant PrPSc was also evident. Further illustrating the biochemical evolution of acutely synaptotoxic species the synaptotoxicity of cM1000 from 30%, 50% and 70% of the TSD, but not at 100% TSD, was abolished by digestion of immuno-captured PrP species with mild PK treatment (5μg/ml for an hour at 37°C), demonstrating that the predominant synaptotoxic PrPSc species up to and including 70% of the TSD were proteinase-sensitive. Overall, these findings in combination with our previous assessments of transmitting prions support that synaptotoxic and infectious M1000 PrPSc species co-exist from at least 30% of the TSD, simultaneously increasing thereafter, albeit with eventual plateauing of transmitting conformers. Although evidence clearly supports that misfolded prion protein (PrPSc) is the principal component of “prions”, underpinning both transmissibility and neurotoxicity, consensus is lacking around the time of appearance and biochemical profile of neurotoxic species during disease evolution. Employing an electrophysiology model, measuring the capacity of brain homogenates derived from across the disease time-course to impair CA1 region long-term potentiation (LTP) and post-tetanic potentiation (PTP) in hippocampal slices, we observed that synaptotoxic species were present from 30% of the terminal stage of disease (TSD). Evidence that synaptotoxicity directly related to PrP species was demonstrated by significant rescue of LTP dysfunction at each time-point through immuno-depleting >~50% of total PrP species from cM1000 preparations. Moreover, size fractionation chromatography revealed that acute synpatotoxicity correlated with predominance of oligomeric PrP species in infected brains across all time points, while additional characterisation of cM1000 demonstrated that the predominant synaptotoxic PrPSc species up to and including 70% of the TSD were quite proteinase-sensitive. These findings in combination with our previous assessments of transmitting prions support that synaptotoxic and infectious M1000 PrPSc species co-exist from at least 30% of the TSD, simultaneously increasing thereafter, with biochemical transformation of synaptotoxic conformers continuing until late in disease.
Collapse
Affiliation(s)
- Simote Totauhelotu Foliaki
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria, Australia
| | - Victoria Lewis
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | | | - Laura Jane Ellett
- Department of Pathology The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| | - Matteo Senesi
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria, Australia
| | | | - Blaine Roberts
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Victoria A. Lawson
- Department of Pathology The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul Anthony Adlard
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Steven John Collins
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
14
|
Vorberg IM. All the Same? The Secret Life of Prion Strains within Their Target Cells. Viruses 2019; 11:v11040334. [PMID: 30970585 DOI: 10.3390/v11040334] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 01/23/2023] Open
Abstract
Prions are infectious β-sheet-rich protein aggregates composed of misfolded prion protein (PrPSc) that do not possess coding nucleic acid. Prions replicate by recruiting and converting normal cellular PrPC into infectious isoforms. In the same host species, prion strains target distinct brain regions and cause different disease phenotypes. Prion strains are associated with biophysically distinct PrPSc conformers, suggesting that strain properties are enciphered within alternative PrPSc quaternary structures. So far it is unknown how prion strains target specific cells and initiate productive infections. Deeper mechanistic insight into the prion life cycle came from cell lines permissive to a range of different prion strains. Still, it is unknown why certain cell lines are refractory to infection by one strain but permissive to another. While pharmacologic and genetic manipulations revealed subcellular compartments involved in prion replication, little is known about strain-specific requirements for endocytic trafficking pathways. This review summarizes our knowledge on how prions replicate within their target cells and on strain-specific differences in prion cell biology.
Collapse
Affiliation(s)
- Ina M Vorberg
- German Center for Neurodegenerative Diseases (DZNE e.V.), Sigmund-Freud-Strasse 27, 53127 Bonn, Germany.
- Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany.
| |
Collapse
|
15
|
Vorberg I, Chiesa R. Experimental models to study prion disease pathogenesis and identify potential therapeutic compounds. Curr Opin Pharmacol 2019; 44:28-38. [PMID: 30878006 DOI: 10.1016/j.coph.2019.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 01/02/2023]
Abstract
Prion diseases are devastating neurodegenerative disorders for which no drugs are available. The successful development of therapeutics depends on drug screening platforms and preclinical models that recapitulate key molecular and pathological features of the disease. Innovative experimental tools have been developed over the last few years that might facilitate drug discovery, including cell-free prion replication assays and prion-infected flies. However, there is still room for improvement. Animal models of genetic prion disease are few, and only partially recapitulate the complexity of the human disorder. Moreover, we still lack a human cell culture model suitable for high-content anti-prion drug screening. This review provides an overview of the models currently used in prion research, and discusses their promise and limitations for drug discovery.
Collapse
Affiliation(s)
- Ina Vorberg
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany.
| | - Roberto Chiesa
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy.
| |
Collapse
|
16
|
Bourkas MEC, Arshad H, Al-Azzawi ZAM, Halgas O, Shikiya RA, Mehrabian M, Schmitt-Ulms G, Bartz JC, Watts JC. Engineering a murine cell line for the stable propagation of hamster prions. J Biol Chem 2019; 294:4911-4923. [PMID: 30705093 DOI: 10.1074/jbc.ra118.007135] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/30/2019] [Indexed: 01/23/2023] Open
Abstract
Prions are infectious protein aggregates that cause several fatal neurodegenerative diseases. Prion research has been hindered by a lack of cellular paradigms for studying the replication of prions from different species. Although hamster prions have been widely used to study prion replication in animals and within in vitro amplification systems, they have proved challenging to propagate in cultured cells. Because the murine catecholaminergic cell line CAD5 is susceptible to a diverse range of mouse prion strains, we hypothesized that it might also be capable of propagating nonmouse prions. Here, using CRISPR/Cas9-mediated genome engineering, we demonstrate that CAD5 cells lacking endogenous mouse PrP expression (CAD5-PrP-/- cells) can be chronically infected with hamster prions following stable expression of hamster PrP. When exposed to the 263K, HY, or 139H hamster prion strains, these cells stably propagated high levels of protease-resistant PrP. Hamster prion replication required absence of mouse PrP, and hamster PrP inhibited the propagation of mouse prions. Cellular homogenates from 263K-infected cells exhibited prion seeding activity in the RT-QuIC assay and were infectious to naïve cells expressing hamster PrP. Interestingly, murine N2a neuroblastoma cells ablated for endogenous PrP expression were susceptible to mouse prions, but not hamster prions upon expression of cognate PrP, suggesting that CAD5 cells either possess cellular factors that enhance or lack factors that restrict the diversity of prion strains that can be propagated. We conclude that transfected CAD5-PrP-/- cells may be a useful tool for assessing the biology of prion strains and dissecting the mechanism of prion replication.
Collapse
Affiliation(s)
- Matthew E C Bourkas
- From the Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada M5T 0S8.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada M5T 0S8
| | - Hamza Arshad
- From the Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada M5T 0S8.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada M5T 0S8
| | - Zaid A M Al-Azzawi
- From the Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada M5T 0S8.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada M5T 0S8
| | - Ondrej Halgas
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada M5T 0S8
| | - Ronald A Shikiya
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, Nebraska, 68178
| | - Mohadeseh Mehrabian
- From the Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada M5T 0S8.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada M5T 0S8, and
| | - Gerold Schmitt-Ulms
- From the Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada M5T 0S8.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada M5T 0S8, and
| | - Jason C Bartz
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, Nebraska, 68178
| | - Joel C Watts
- From the Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada M5T 0S8, .,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada M5T 0S8
| |
Collapse
|
17
|
Abstract
The development of multiple cell culture models of prion infection over the last two decades has led to a significant increase in our understanding of how prions infect cells. In particular, new techniques to distinguish exogenous from endogenous prions have allowed us for the first time to look in depth at the earliest stages of prion infection through to the establishment of persistent infection. These studies have shown that prions can infect multiple cell types, both neuronal and nonneuronal. Once in contact with the cell, they are rapidly taken up via multiple endocytic pathways. After uptake, the initial replication of prions occurs almost immediately on the plasma membrane and within multiple endocytic compartments. Following this acute stage of prion replication, persistent prion infection may or may not be established. Establishment of a persistent prion infection in cells appears to depend upon the achievement of a delicate balance between the rate of prion replication and degradation, the rate of cell division, and the efficiency of prion spread from cell to cell. Overall, cell culture models have shown that prion infection of the cell is a complex and variable process which can involve multiple cellular pathways and compartments even within a single cell.
Collapse
Affiliation(s)
- Suzette A Priola
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States.
| |
Collapse
|
18
|
Prion acute synaptotoxicity is largely driven by protease-resistant PrPSc species. PLoS Pathog 2018; 14:e1007214. [PMID: 30089152 PMCID: PMC6101418 DOI: 10.1371/journal.ppat.1007214] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/20/2018] [Accepted: 07/12/2018] [Indexed: 01/09/2023] Open
Abstract
Although misfolding of normal prion protein (PrPC) into abnormal conformers (PrPSc) is critical for prion disease pathogenesis our current understanding of the underlying molecular pathophysiology is rudimentary. Exploiting an electrophysiology paradigm, herein we report that at least modestly proteinase K (PK)-resistant PrPSc (PrPres) species are acutely synaptotoxic. Brief exposure to ex vivo PrPSc from two mouse-adapted prion strains (M1000 and MU02) prepared as crude brain homogenates (cM1000 and cMU02) and cell lysates from chronically M1000-infected RK13 cells (MoRK13-Inf) caused significant impairment of hippocampal CA1 region long-term potentiation (LTP), with the LTP disruption approximating that reported during the evolution of murine prion disease. Proof of PrPSc (especially PrPres) species as the synaptotoxic agent was demonstrated by: significant rescue of LTP following selective immuno-depletion of total PrP from cM1000 (dM1000); modestly PK-treated cM1000 (PK+M1000) retaining full synaptotoxicity; and restoration of the LTP impairment when employing reconstituted, PK-eluted, immuno-precipitated M1000 preparations (PK+IP-M1000). Additional detailed electrophysiological analyses exemplified by impairment of post-tetanic potentiation (PTP) suggest possible heightened pre-synaptic vulnerability to the acute synaptotoxicity. This dysfunction correlated with cumulative insufficiency of replenishment of the readily releasable pool (RRP) of vesicles during repeated high-frequency stimulation utilised for induction of LTP. Broadly comparable results with LTP and PTP impairment were obtained utilizing hippocampal slices from PrPC knockout (PrPo/o) mice, with cM1000 serial dilution assessments revealing similar sensitivity of PrPo/o and wild type (WT) slices. Size fractionation chromatography demonstrated that synaptotoxic PrP correlated with PK-resistant species >100kDa, consistent with multimeric PrPSc, with levels of these species >6 ng/ml appearing sufficient to induce synaptic dysfunction. Biochemical analyses of hippocampal slices manifesting acute synaptotoxicity demonstrated reduced levels of multiple key synaptic proteins, albeit with noteworthy differences in PrPo/o slices, while such changes were absent in hippocampi demonstrating rescued LTP through treatment with dM1000. Our findings offer important new mechanistic insights into the synaptic impairment underlying prion disease, enhancing prospects for development of targeted effective therapies. Misfolding of the normal prion protein (PrPC) into disease-associated conformations (PrPSc) is the critical initiating step for prion diseases. Similar to other neurodegenerative disorders, progressive failure of brain synapses is considered a primary deleterious event underpinning prion disease evolution. Our current understanding of the underlying mechanisms associated with synaptic failure is rudimentary contributing to difficulties in developing effective treatments. Herein we report the use of an electrophysiology paradigm that allowed us to demonstrate that at least modestly proteinase K (PK)-resistant PrPSc species from two mouse-adapted prion strains (M1000 and MU02) are directly synaptotoxic causing significant acute impairment of hippocampal CA1 region long-term potentiation (LTP). Of note, the LTP disruption approximated that reported in prion animal models. Additional detailed analyses provided novel pathophysiological insights suggesting possible heightened pre-synaptic vulnerability to the acute synaptotoxicity through impairment of replenishment of the readily releasable pool of neurotransmitter vesicles, while biochemical analyses demonstrated reduced levels of multiple key pre-and post-synaptic proteins. Broadly similar acute synaptic dysfunction and dose-response susceptibility were observed in slices from mice not expressing PrPC albeit with minor but noteworthy differences in electrophysiological and biochemical findings. Our study offers important new mechanistic insights into the synaptic impairment underlying prion disease, enhancing prospects for development effective therapies.
Collapse
|
19
|
Krejciova Z, Alibhai J, Zhao C, Krencik R, Rzechorzek NM, Ullian EM, Manson J, Ironside JW, Head MW, Chandran S. Human stem cell-derived astrocytes replicate human prions in a PRNP genotype-dependent manner. J Exp Med 2017; 214:3481-3495. [PMID: 29141869 PMCID: PMC5716027 DOI: 10.1084/jem.20161547] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 08/07/2017] [Accepted: 09/27/2017] [Indexed: 01/09/2023] Open
Abstract
Prions are infectious agents that cause neurodegenerative diseases such as Creutzfeldt-Jakob disease (CJD). The absence of a human cell culture model that replicates human prions has hampered prion disease research for decades. In this paper, we show that astrocytes derived from human induced pluripotent stem cells (iPSCs) support the replication of prions from brain samples of CJD patients. For experimental exposure of astrocytes to variant CJD (vCJD), the kinetics of prion replication occur in a prion protein codon 129 genotype-dependent manner, reflecting the genotype-dependent susceptibility to clinical vCJD found in patients. Furthermore, iPSC-derived astrocytes can replicate prions associated with the major sporadic CJD strains found in human patients. Lastly, we demonstrate the subpassage of prions from infected to naive astrocyte cultures, indicating the generation of prion infectivity in vitro. Our study addresses a long-standing gap in the repertoire of human prion disease research, providing a new in vitro system for accelerated mechanistic studies and drug discovery.
Collapse
Affiliation(s)
- Zuzana Krejciova
- National CJD Research & Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK,Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA
| | - James Alibhai
- National CJD Research & Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Chen Zhao
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Robert Krencik
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX
| | - Nina M. Rzechorzek
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK,Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Edinburgh, Scotland, UK
| | - Erik M. Ullian
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA
| | - Jean Manson
- Neurobiology Division, The Roslin Institute, University of Edinburgh, Edinburgh, Scotland, UK
| | - James W. Ironside
- National CJD Research & Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Mark W. Head
- National CJD Research & Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Siddharthan Chandran
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland, UK,UK Dementia Research Institute, University of Edinburgh, Edinburgh, Scotland, UK,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore, India,Correspondence to Siddharthan Chandran:
| |
Collapse
|
20
|
Tamgüney G, Korczyn AD. A critical review of the prion hypothesis of human synucleinopathies. Cell Tissue Res 2017; 373:213-220. [DOI: 10.1007/s00441-017-2712-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/11/2017] [Indexed: 01/01/2023]
|
21
|
Dassanayake RP, Zhuang D, Truscott TC, Madsen-Bouterse SA, O'Rourke KI, Schneider DA. A transfectant RK13 cell line permissive to classical caprine scrapie prion propagation. Prion 2017; 10:153-64. [PMID: 27216989 DOI: 10.1080/19336896.2016.1166324] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
To assess scrapie infectivity associated with caprine-origin tissues, bioassay can be performed using kids, lambs or transgenic mice expressing caprine or ovine prion (PRNP) alleles, but the incubation periods are fairly long. Although several classical ovine scrapie prion permissive cell lines with the ability to detect brain-derived scrapie prion have been available, no classical caprine scrapie permissive cell line is currently available. Therefore, the aims of this study were to generate a rabbit kidney epithelial cell line (RK13) stably expressing caprine wild-type PRNP (cpRK13) and then to assess permissiveness of cpRK13 cells to classical caprine scrapie prion propagation. The cpRK13 and plasmid control RK13 (pcRK13) cells were incubated with brain-derived classical caprine scrapie inocula prepared from goats or ovinized transgenic mice (Tg338, express ovine VRQ allele) infected with caprine scrapie. Significant PrP(Sc) accumulation, which is indicative of scrapie prion propagation, was detected by TSE ELISA and immunohistochemistry in cpRK13 cells inoculated with classical caprine scrapie inocula. Western blot analysis revealed the typical proteinase K-resistant 3 PrP(res) isoforms in the caprine scrapie prion inoculated cpRK13 cell lysate. Importantly, PrP(Sc) accumulation was not detected in similarly inoculated pcRK13 cells, whether by TSE ELISA, immunohistochemistry, or western blot. These findings suggest that caprine scrapie prions can be propagated in cpRK13 cells, thus this cell line may be a useful tool for the assessment of classical caprine prions in the brain tissues of goats.
Collapse
Affiliation(s)
- Rohana P Dassanayake
- a Department of Veterinary Microbiology and Pathology , College of Veterinary Medicine, Washington State University , Pullman , WA , USA
| | - Dongyue Zhuang
- b Animal Disease Research Unit, Agricultural Research Service , U.S. Department of Agriculture , Pullman , WA , USA
| | - Thomas C Truscott
- b Animal Disease Research Unit, Agricultural Research Service , U.S. Department of Agriculture , Pullman , WA , USA
| | - Sally A Madsen-Bouterse
- a Department of Veterinary Microbiology and Pathology , College of Veterinary Medicine, Washington State University , Pullman , WA , USA
| | - Katherine I O'Rourke
- a Department of Veterinary Microbiology and Pathology , College of Veterinary Medicine, Washington State University , Pullman , WA , USA
| | - David A Schneider
- a Department of Veterinary Microbiology and Pathology , College of Veterinary Medicine, Washington State University , Pullman , WA , USA ;,b Animal Disease Research Unit, Agricultural Research Service , U.S. Department of Agriculture , Pullman , WA , USA
| |
Collapse
|
22
|
Vella LJ, Coleman B, Hill AF. Generation of Infectious Prions and Detection with the Prion-Infected Cell Assay. Methods Mol Biol 2017; 1658:105-118. [PMID: 28861786 DOI: 10.1007/978-1-4939-7244-9_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Cell lines propagating prions are an efficient and useful means for studying the cellular and molecular mechanisms implicated in prion disease. Utilization of cell-based models has led to the finding that PrPC and PrPSc are released from cells in association with extracellular vesicles known as exosomes. Exosomes have been shown to act as vehicles for infectivity, transferring infectivity between cell lines and providing a mechanism for prion spread between tissues. Here, we describe the methods for generating a prion-propagating cell line with prion-infected brain homogenate, cell lysate, conditioned media, and exosomes and also detection of protease-resistant PrP with the prion-infected cell assay.
Collapse
Affiliation(s)
- Laura J Vella
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Bradley Coleman
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Andrew F Hill
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia. .,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia.
| |
Collapse
|
23
|
Abstract
Like numerous proteins of various structural and functional classes, the glycosylphosphatidylinositol (GPI)-anchored cellular prion protein (PrPC) has been recognized to undergo endoproteolytic processing for decades, a phenomenon observed in various cultured cell lines, as well as human and several animal tissue extracts. Despite this, the physiological significance of PrPC proteolytic cleavage has not yet been entirely elucidated. Experimental evidence suggests independent normal biological functions of the full-length and truncated PrPC species, as well as probable links of endoproteolysis to prion disease transmission susceptibility, pathogenesis, and toxicity. The accurate characterization of constitutive PrPC processing, through the method outlined in this chapter, is therefore an important tool in order to investigate the biological relevance of the alternative cleavage events.
Collapse
Affiliation(s)
- Victoria Lewis
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
24
|
Abstract
Prion diseases are a group of invariably fatal and transmissible neurodegenerative disorders that are associated with the misfolding of the normal cellular prion protein, with the misfolded conformers constituting an infectious unit referred to as a "prion". Prions can spread within an affected organism by directly propagating this misfolding within and between cells and can transmit disease between animals of the same and different species. Prion diseases have a range of clinical phenotypes in humans and animals, with a principle determinant of this attributed to different conformations of the misfolded protein, referred to as prion strains. This chapter will describe the different clinical manifestations of prion diseases, the evidence that these diseases can be transmitted by an infectious protein and how the misfolding of this protein causes disease.
Collapse
|
25
|
Arellano-Anaya ZE, Huor A, Leblanc P, Andréoletti O, Vilette D. Expression of Heterologous PrP and Prion Propagation in RK13 Cells. Methods Mol Biol 2017; 1658:95-104. [PMID: 28861785 DOI: 10.1007/978-1-4939-7244-9_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cultured cells are valuable models to study prion infections at the cellular level. Unfortunately, the vast majority of cell lines are resistant to the propagation of prion agents. The rabbit epithelial RK13 cell line is among the few cell lines permissive to prion infection. When genetically engineered to express heterologous PrP proteins, RK13 cells become permissive to several strains of prions from various animal species. Here, we describe the generation of stable RK13 cell clones expressing a heterologous PrP protein in an inducible manner, the establishment and maintenance of chronically infected cultures, and the selection of cell clones suitable for cell-based titration of prions.
Collapse
Affiliation(s)
- Zaira E Arellano-Anaya
- INRA, UMR 1225, IHAP, 31076, Toulouse, France
- Université de Toulouse, INP, ENVT, UMR1225, IHAP, 31076, Toulouse, France
| | - Alvina Huor
- INRA, UMR 1225, IHAP, 31076, Toulouse, France
- Université de Toulouse, INP, ENVT, UMR1225, IHAP, 31076, Toulouse, France
| | - Pascal Leblanc
- CNRS, UMR5239, Laboratoire de Biologie Moléculaire de la Cellule (LBMC), Equipe Différenciation Neuromusculaire, Ecole Normale Supérieure-Lyon, 46 allée d'Italie, 69364, Lyon Cedex 07, France
| | - Olivier Andréoletti
- INRA, UMR 1225, IHAP, 31076, Toulouse, France
- Université de Toulouse, INP, ENVT, UMR1225, IHAP, 31076, Toulouse, France
| | - Didier Vilette
- INRA, UMR 1225, IHAP, 31076, Toulouse, France.
- Université de Toulouse, INP, ENVT, UMR1225, IHAP, 31076, Toulouse, France.
| |
Collapse
|
26
|
Collins SJ, Haigh CL. Simplified Murine 3D Neuronal Cultures for Investigating Neuronal Activity and Neurodegeneration. Cell Biochem Biophys 2016; 75:3-13. [PMID: 27796787 DOI: 10.1007/s12013-016-0768-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 10/17/2016] [Indexed: 12/28/2022]
Abstract
The ability to model brain tissue in three-dimensions offers new potential for elucidating functional cellular interactions and corruption of such functions during pathogenesis. Many protocols now exist for growing neurones in three-dimensions and these vary in complexity and cost. Herein, we describe a straight-forward method for generating three-dimensional, terminally differentiated central nervous system cultures from adult murine neural stem cells. The protocol requires no specialist equipment, is not labour intensive or expensive and produces mature cultures within 10 days that can survive beyond a month. Populations of functional glutamatergic neurones could be identified within cultures. Additionally, the three dimensional neuronal cultures can be used to investigate tissue changes during the development of neurodegenerative disease where demonstration of hallmark features, such as plaque generation, has not previously been possible using two-dimensional cultures of neuronal cells. Using a prion model of acquired neurodegenerative disease, biochemical changes indicative of prion pathology were induced within 2-3 weeks in the three dimensional cultures. Our findings show that tissue differentiated in this simplified three dimensional culture model is physiologically competent to model central nervous system cellular behaviour as well as manifest the functional failures and pathological changes associated with neurodegenerative disease.
Collapse
Affiliation(s)
- Steven J Collins
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, The University of Melbourne, 30 Royal Parade, Parkville, Melbourne, VIC, 3010, Australia
| | - Cathryn L Haigh
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, The University of Melbourne, 30 Royal Parade, Parkville, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
27
|
Generating Bona Fide Mammalian Prions with Internal Deletions. J Virol 2016; 90:6963-6975. [PMID: 27226369 DOI: 10.1128/jvi.00555-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 05/14/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Mammalian prions are PrP proteins with altered structures causing transmissible fatal neurodegenerative diseases. They are self-perpetuating through formation of beta-sheet-rich assemblies that seed conformational change of cellular PrP. Pathological PrP usually forms an insoluble protease-resistant core exhibiting beta-sheet structures but no more alpha-helical content, loosing the three alpha-helices contained in the correctly folded PrP. The lack of a high-resolution prion structure makes it difficult to understand the dynamics of conversion and to identify elements of the protein involved in this process. To determine whether completeness of residues within the protease-resistant domain is required for prions, we performed serial deletions in the helix H2 C terminus of ovine PrP, since this region has previously shown some tolerance to sequence changes without preventing prion replication. Deletions of either four or five residues essentially preserved the overall PrP structure and mutant PrP expressed in RK13 cells were efficiently converted into bona fide prions upon challenge by three different prion strains. Remarkably, deletions in PrP facilitated the replication of two strains that otherwise do not replicate in this cellular context. Prions with internal deletion were self-propagating and de novo infectious for naive homologous and wild-type PrP-expressing cells. Moreover, they caused transmissible spongiform encephalopathies in mice, with similar biochemical signatures and neuropathologies other than the original strains. Prion convertibility and transfer of strain-specific information are thus preserved despite shortening of an alpha-helix in PrP and removal of residues within prions. These findings provide new insights into sequence/structure/infectivity relationship for prions. IMPORTANCE Prions are misfolded PrP proteins that convert the normal protein into a replicate of their own abnormal form. They are responsible for invariably fatal neurodegenerative disorders. Other aggregation-prone proteins appear to have a prion-like mode of expansion in brains, such as in Alzheimer's or Parkinson's diseases. To date, the resolution of prion structure remains elusive. Thus, to genetically define the landscape of regions critical for prion conversion, we tested the effect of short deletions. We found that, surprisingly, removal of a portion of PrP, the C terminus of alpha-helix H2, did not hamper prion formation but generated infectious agents with an internal deletion that showed characteristics essentially similar to those of original infecting strains. Thus, we demonstrate that completeness of the residues inside prions is not necessary for maintaining infectivity and the main strain-specific information, while reporting one of the few if not the only bona fide prions with an internal deletion.
Collapse
|
28
|
Guo BB, Bellingham SA, Hill AF. Stimulating the Release of Exosomes Increases the Intercellular Transfer of Prions. J Biol Chem 2016; 291:5128-37. [PMID: 26769968 DOI: 10.1074/jbc.m115.684258] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Indexed: 01/20/2023] Open
Abstract
Exosomes are small extracellular vesicles released by cells and play important roles in intercellular communication and pathogen transfer. Exosomes have been implicated in several neurodegenerative diseases, including prion disease and Alzheimer disease. Prion disease arises upon misfolding of the normal cellular prion protein, PrP(C), into the disease-associated isoform, PrP(Sc). The disease has a unique transmissible etiology, and exosomes represent a novel and efficient method for prion transmission. The precise mechanism by which prions are transmitted from cell to cell remains to be fully elucidated, although three hypotheses have been proposed: direct cell-cell contact, tunneling nanotubes, and exosomes. Given the reported presence of exosomes in biological fluids and in the lipid and nucleic acid contents of exosomes, these vesicles represent an ideal mechanism for encapsulating prions and potential cofactors to facilitate prion transmission. This study investigates the relationship between exosome release and intercellular prion dissemination. Stimulation of exosome release through treatment with an ionophore, monensin, revealed a corresponding increase in intercellular transfer of prion infectivity. Conversely, inhibition of exosome release using GW4869 to target the neutral sphingomyelinase pathway induced a decrease in intercellular prion transmission. Further examination of the effect of monensin on PrP conversion revealed that monensin also alters the conformational stability of PrP(C), leading to increased generation of proteinase K-resistant prion protein. The findings presented here provide support for a positive relationship between exosome release and intercellular transfer of prion infectivity, highlighting an integral role for exosomes in facilitating the unique transmissible nature of prions.
Collapse
Affiliation(s)
- Belinda B Guo
- From the Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and
| | - Shayne A Bellingham
- From the Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and
| | - Andrew F Hill
- From the Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia and the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| |
Collapse
|
29
|
Haigh CL, Drew SC. Cavitation during the protein misfolding cyclic amplification (PMCA) method – The trigger for de novo prion generation? Biochem Biophys Res Commun 2015; 461:494-500. [DOI: 10.1016/j.bbrc.2015.04.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/07/2015] [Indexed: 12/14/2022]
|
30
|
Ellett LJ, Coleman BM, Shambrook MC, Johanssen VA, Collins SJ, Masters CL, Hill AF, Lawson VA. Glycosaminoglycan sulfation determines the biochemical properties of prion protein aggregates. Glycobiology 2015; 25:745-55. [PMID: 25701659 DOI: 10.1093/glycob/cwv014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 02/15/2015] [Indexed: 02/06/2023] Open
Abstract
Prion diseases are transmissible neurodegenerative disorders associated with the conversion of the cellular prion protein, PrP(C), to a misfolded isoform called PrP(Sc). Although PrP(Sc) is a necessary component of the infectious prion, additional factors, or cofactors, have been shown to contribute to the efficient formation of transmissible PrP(Sc). Glycosaminoglycans (GAGs) are attractive cofactor candidates as they can be found associated with PrP(Sc) deposits, have been shown to enhance PrP misfolding in vitro, are found in the same cellular compartments as PrP(C) and have been shown to be disease modifying in vivo. Here we investigated the effects of the sulfated GAGs, heparin and heparan sulfate (HS), on disease associated misfolding of full-length recombinant PrP. More specifically, the degree of sulfation of these molecules was investigated for its role in modulating the disease-associated characteristics of PrP. Both heparin and HS induced a β-sheet conformation in recombinant PrP that was associated with the formation of aggregated species; however, the biochemical properties of the aggregates formed in the presence of heparin or HS varied in solubility and protease resistance. Furthermore, these properties could be modified by changes in GAG sulfation, indicating that subtle changes in the properties of prion disease cofactors could initiate disease associated misfolding.
Collapse
Affiliation(s)
| | - Bradley M Coleman
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute
| | - Mitch C Shambrook
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute
| | | | | | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University Of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew F Hill
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute
| | | |
Collapse
|
31
|
The standard scrapie cell assay: development, utility and prospects. Viruses 2015; 7:180-98. [PMID: 25602372 PMCID: PMC4306833 DOI: 10.3390/v7010180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/06/2015] [Indexed: 11/23/2022] Open
Abstract
Prion diseases are a family of fatal neurodegenerative diseases that involve the misfolding of a host protein, PrPC. Measuring prion infectivity is necessary for determining efficacy of a treatment or infectivity of a prion purification procedure; animal bioassays are, however, very expensive and time consuming. The Standard Scrapie Cell Assay (SSCA) provides an alternative approach. The SSCA facilitates quantitative in vitro analysis of prion strains, titres and biological properties. Given its robust nature and potential for high throughput, the SSCA has substantial utility for in vitro characterization of prions and can be deployed in a number of settings. Here we provide an overview on establishing the SSCA, its use in studies of disease dissemination and pathogenesis, potential pitfalls and a number of remaining challenges.
Collapse
|
32
|
Guo BB, Bellingham SA, Hill AF. The neutral sphingomyelinase pathway regulates packaging of the prion protein into exosomes. J Biol Chem 2014; 290:3455-67. [PMID: 25505180 DOI: 10.1074/jbc.m114.605253] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Prion diseases are a group of transmissible, fatal neurodegenerative disorders associated with the misfolding of the host-encoded prion protein, PrP(C), into a disease-associated form, PrP(Sc). The transmissible prion agent is principally formed of PrP(Sc) itself and is associated with extracellular vesicles known as exosomes. Exosomes are released from cells both in vitro and in vivo, and have been proposed as a mechanism by which prions spread intercellularly. The biogenesis of exosomes occurs within the endosomal system, through formation of intraluminal vesicles (ILVs), which are subsequently released from cells as exosomes. ILV formation is known to be regulated by the endosomal sorting complexes required for transport (ESCRT) machinery, although an alternative neutral sphingomyelinase (nSMase) pathway has been suggested to also regulate this process. Here, we investigate a role for the nSMase pathway in exosome biogenesis and packaging of PrP into these vesicles. Inhibition of the nSMase pathway using GW4869 revealed a role for the nSMase pathway in both exosome formation and PrP packaging. In agreement, targeted knockdown of nSMase1 and nSMase2 in mouse neurons using lentivirus-mediated RNAi also decreases exosome release, demonstrating the nSMase pathway regulates the biogenesis and release of exosomes. We also demonstrate that PrP(C) packaging is dependent on nSMase2, whereas the packaging of disease-associated PrP(Sc) into exosomes occurs independently of nSMase2. These findings provide further insight into prion transmission and identify a pathway which directly assists exosome-mediated transmission of prions.
Collapse
Affiliation(s)
- Belinda B Guo
- From the Department of Biochemistry and Molecular Biology, The University of Melbourne, VIC 3010, Australia and the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, VIC 3010, Australia
| | - Shayne A Bellingham
- From the Department of Biochemistry and Molecular Biology, The University of Melbourne, VIC 3010, Australia and the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, VIC 3010, Australia
| | - Andrew F Hill
- From the Department of Biochemistry and Molecular Biology, The University of Melbourne, VIC 3010, Australia and the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, VIC 3010, Australia
| |
Collapse
|
33
|
Mediano DR, Sanz-Rubio D, Ranera B, Bolea R, Martín-Burriel I. The potential of mesenchymal stem cell in prion research. Zoonoses Public Health 2014; 62:165-78. [PMID: 24854140 DOI: 10.1111/zph.12138] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Indexed: 01/09/2023]
Abstract
Scrapie and bovine spongiform encephalopathy are fatal neurodegenerative diseases caused by the accumulation of a misfolded protein (PrP(res)), the pathological form of the cellular prion protein (PrP(C)). For the last decades, prion research has greatly progressed, but many questions need to be solved about prion replication mechanisms, cell toxicity, differences in genetic susceptibility, species barrier or the nature of prion strains. These studies can be developed in murine models of transmissible spongiform encephalopathies, although development of cell models for prion replication and sample titration could reduce economic and timing costs and also serve for basic research and treatment testing. Some murine cell lines can replicate scrapie strains previously adapted in mice and very few show the toxic effects of prion accumulation. Brain cell primary cultures can be more accurate models but are difficult to develop in naturally susceptible species like humans or domestic ruminants. Stem cells can be differentiated into neuron-like cells and be infected by prions. However, the use of embryo stem cells causes ethical problems in humans. Mesenchymal stem cells (MSCs) can be isolated from many adult tissues, including bone marrow, adipose tissue or even peripheral blood. These cells differentiate into neuronal cells, express PrP(C) and can be infected by prions in vitro. In addition, in the last years, these cells are being used to develop therapies for many diseases, including neurodegenerative diseases. We review here the use of cell models in prion research with a special interest in the potential use of MSCs.
Collapse
Affiliation(s)
- D R Mediano
- Facultad de Veterinaria, Laboratorio de Genética Bioquímica, Universidad de Zaragoza, Zaragoza, Spain
| | | | | | | | | |
Collapse
|
34
|
Krejciova Z, De Sousa P, Manson J, Ironside JW, Head MW. Human tonsil-derived follicular dendritic-like cells are refractory to human prion infection in vitro and traffic disease-associated prion protein to lysosomes. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:64-70. [PMID: 24183781 PMCID: PMC3873479 DOI: 10.1016/j.ajpath.2013.09.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 09/12/2013] [Accepted: 09/16/2013] [Indexed: 01/09/2023]
Abstract
The molecular mechanisms involved in human cellular susceptibility to prion infection remain poorly defined. This is due, in part, to the absence of any well characterized and relevant cultured human cells susceptible to infection with human prions, such as those involved in Creutzfeldt-Jakob disease. In variant Creutzfeldt-Jakob disease, prion replication is thought to occur first in the lymphoreticular system and then spread into the brain. We have, therefore, examined the susceptibility of a human tonsil-derived follicular dendritic cell-like cell line (HK) to prion infection. HK cells were found to display a readily detectable, time-dependent increase in cell-associated abnormal prion protein (PrP(TSE)) when exposed to medium spiked with Creutzfeldt-Jakob disease brain homogenate, resulting in a coarse granular perinuclear PrP(TSE) staining pattern. Despite their high level of cellular prion protein expression, HK cells failed to support infection, as judged by longer term maintenance of PrP(TSE) accumulation. Colocalization studies revealed that exposure of HK cells to brain homogenate resulted in increased numbers of detectable lysosomes and that these structures immunostained intensely for PrP(TSE) after exposure to Creutzfeldt-Jakob disease brain homogenate. Our data suggest that human follicular dendritic-like cells and perhaps other human cell types are able to avoid prion infection by efficient lysosomal degradation of PrP(TSE).
Collapse
Affiliation(s)
- Zuzana Krejciova
- National Creutzfeldt-Jakob Disease Research & Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Paul De Sousa
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Jean Manson
- Neurobiology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, United Kingdom
| | - James W Ironside
- National Creutzfeldt-Jakob Disease Research & Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark W Head
- National Creutzfeldt-Jakob Disease Research & Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
35
|
Pathogenic mutations within the hydrophobic domain of the prion protein lead to the formation of protease-sensitive prion species with increased lethality. J Virol 2013; 88:2690-703. [PMID: 24352465 DOI: 10.1128/jvi.02720-13] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
UNLABELLED Prion diseases are a group of fatal and incurable neurodegenerative diseases affecting both humans and animals. The principal mechanism of these diseases involves the misfolding the host-encoded cellular prion protein, PrP(C), into the disease-associated isoform, PrP(Sc). Familial forms of human prion disease include those associated with the mutations G114V and A117V, which lie in the hydrophobic domain of PrP. Here we have studied the murine homologues (G113V and A116V) of these mutations using cell-based and animal models of prion infection. Under normal circumstances, the mutant forms of PrP(C) share similar processing, cellular localization, and physicochemical properties with wild-type mouse PrP (MoPrP). However, upon exposure of susceptible cell lines expressing these mutants to infectious prions, very low levels of protease-resistant aggregated PrP(Sc) are formed. Subsequent mouse bioassay revealed high levels of infectivity present in these cells. Thus, these mutations appear to limit the formation of aggregated PrP(Sc), giving rise to the accumulation of a relatively soluble, protease sensitive, prion species that is highly neurotoxic. Given that these mutations lie next to the glycine-rich region of PrP that can abrogate prion infection, these findings provide further support for small, protease-sensitive prion species having a significant role in the progression of prion disease and that the hydrophobic domain is an important determinant of PrP conversion. IMPORTANCE Prion diseases are transmissible neurodegenerative diseases associated with an infectious agent called a prion. Prions are comprised of an abnormally folded form of the prion protein (PrP) that is normally resistant to enzymes called proteases. In humans, prion disease can occur in individuals who inherited mutations in the prion protein gene. Here we have studied the effects of two of these mutations and show that they influence the properties of the prions that can be formed. We show that the mutants make highly infectious prions that are more sensitive to protease treatment. This study highlights a certain region of the prion protein as being involved in this effect and demonstrates that prions are not always resistant to protease treatment.
Collapse
|
36
|
Cui HL, Guo B, Scicluna B, Coleman BM, Lawson VA, Ellett L, Meikle PJ, Bukrinsky M, Mukhamedova N, Sviridov D, Hill AF. Prion infection impairs cholesterol metabolism in neuronal cells. J Biol Chem 2013; 289:789-802. [PMID: 24280226 PMCID: PMC3887205 DOI: 10.1074/jbc.m113.535807] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Conversion of prion protein (PrP(C)) into a pathological isoform (PrP(Sc)) during prion infection occurs in lipid rafts and is dependent on cholesterol. Here, we show that prion infection increases the abundance of cholesterol transporter, ATP-binding cassette transporter type A1 (ATP-binding cassette transporter type A1), but reduces cholesterol efflux from neuronal cells leading to the accumulation of cellular cholesterol. Increased abundance of ABCA1 in prion disease was confirmed in prion-infected mice. Mechanistically, conversion of PrP(C) to the pathological isoform led to PrP(Sc) accumulation in rafts, displacement of ABCA1 from rafts and the cell surface, and enhanced internalization of ABCA1. These effects were abolished with reversal of prion infection or by loading cells with cholesterol. Stimulation of ABCA1 expression with liver X receptor agonist or overexpression of heterologous ABCA1 reduced the conversion of prion protein into the pathological form upon infection. These findings demonstrate a reciprocal connection between prion infection and cellular cholesterol metabolism, which plays an important role in the pathogenesis of prion infection in neuronal cells.
Collapse
Affiliation(s)
- Huanhuan L Cui
- From the Baker Heart and Diabetes Institute, Melbourne, Victoria 8008, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Infectious prions accumulate to high levels in non proliferative C2C12 myotubes. PLoS Pathog 2013; 9:e1003755. [PMID: 24244171 PMCID: PMC3820720 DOI: 10.1371/journal.ppat.1003755] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 09/25/2013] [Indexed: 02/05/2023] Open
Abstract
Prion diseases are driven by the strain-specific, template-dependent transconformation of the normal cellular prion protein (PrPC) into a disease specific isoform PrPSc. Cell culture models of prion infection generally use replicating cells resulting in lower levels of prion accumulation compared to animals. Using non-replicating cells allows the accumulation of higher levels of PrPSc and, thus, greater amounts of infectivity. Here, we infect non-proliferating muscle fiber myotube cultures prepared from differentiated myoblasts. We demonstrate that prion-infected myotubes generate substantial amounts of PrPSc and that the level of infectivity produced in these post-mitotic cells, 105.5 L.D.50/mg of total protein, approaches that observed in vivo. Exposure of the myotubes to different mouse-adapted agents demonstrates strain-specific replication of infectious agents. Mouse-derived myotubes could not be infected with hamster prions suggesting that the species barrier effect is intact. We suggest that non-proliferating myotubes will be a valuable model system for generating infectious prions and for screening compounds for anti-prion activity. This manuscript describes the generation of a new cell culture system to study the replication of infectious prions. While numerous cell lines exist that can replicate prions, these systems are usually based upon proliferating cells. As mammalian cell cultures double approximately every day, prions established in the culture must also, at least, double to be maintained. This is problematic, however, as prions replicate relatively slowly and cell replication may outpace prion replication. In fact, many cell culture systems do not replicate prions and those that do often do not replicate all strains of prions. Here we describe the use of differentiated non-proliferative muscle cells to replicate prions without the interfering effect of cell division. We observed that prions accumulate to very high levels in this muscle cell culture with infectivity approaching that observed in animals.
Collapse
|
38
|
Molecular dynamics studies on the NMR and X-ray structures of rabbit prion proteins. J Theor Biol 2013; 342:70-82. [PMID: 24184221 DOI: 10.1016/j.jtbi.2013.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 08/25/2013] [Accepted: 10/09/2013] [Indexed: 12/27/2022]
Abstract
Prion diseases, traditionally referred to as transmissible spongiform encephalopathies (TSEs), are invariably fatal and highly infectious neurodegenerative diseases that affect a wide variety of mammalian species, manifesting as scrapie in sheep and goats, bovine spongiform encephalopathy (BSE or mad-cow disease) in cattle, chronic wasting disease in deer and elk, and Creutzfeldt-Jakob diseases, Gerstmann-Sträussler-Scheinker syndrome, fatal familial insomnia, and kulu in humans, etc. These neurodegenerative diseases are caused by the conversion from a soluble normal cellular prion protein (PrP(C)) into insoluble abnormally folded infectious prions (PrP(Sc)), and the conversion of PrP(C) to PrP(Sc) is believed to involve conformational change from a predominantly α-helical protein to one rich in β-sheet structure. Such a conformational change may be amenable to study by molecular dynamics (MD) techniques. For rabbits, classical studies show that they have a low susceptibility to be infected by PrP(Sc), but recently it was reported that rabbit prions can be generated through saPMCA (serial automated Protein Misfolding Cyclic Amplification) in vitro and the rabbit prion is infectious and transmissible. In this paper, we first do a detailed survey on the research advances of rabbit prion protein (RaPrP) and then we perform MD simulations on the NMR and X-ray molecular structures of rabbit prion protein wild-type and mutants. The survey shows to us that rabbits were not challenged directly in vivo with other known prion strains and the saPMCA result did not pass the test of the known BSE strain of cattle. Thus, we might still look rabbits as a prion resistant species. MD results indicate that the three α-helices of the wild-type are stable under the neutral pH environment (but under low pH environment the three α-helices have been unfolded into β-sheets), and the three α-helices of the mutants (I214V and S173N) are unfolded into rich β-sheet structures under the same pH environment. In addition, we found an interesting result that the salt bridges such as ASP201-ARG155, ASP177-ARG163 contribute greatly to the structural stability of RaPrP.
Collapse
|
39
|
Sinclair L, Lewis V, Collins SJ, Haigh CL. Cytosolic caspases mediate mislocalised SOD2 depletion in an in vitro model of chronic prion infection. Dis Model Mech 2013; 6:952-63. [PMID: 23580200 PMCID: PMC3701215 DOI: 10.1242/dmm.010678] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Oxidative stress as a contributor to neuronal death during prion infection is supported by the fact that various oxidative damage markers accumulate in the brain during the course of this disease. The normal cellular substrate of the causative agent, the prion protein, is also linked with protective functions against oxidative stress. Our previous work has found that, in chronic prion infection, an apoptotic subpopulation of cells exhibit oxidative stress and the accumulation of oxidised lipid and protein aggregates with caspase recruitment. Given the likely failure of antioxidant defence mechanisms within apoptotic prion-infected cells, we aimed to investigate the role of the crucial antioxidant pathway components, superoxide dismutases (SOD) 1 and 2, in an in vitro model of chronic prion infection. Increased total SOD activity, attributable to SOD1, was found in the overall population coincident with a decrease in SOD2 protein levels. When apoptotic cells were separated from the total population, the induction of SOD activity in the infected apoptotic cells was lost, with activity reduced back to levels seen in mock-infected control cells. In addition, mitochondrial superoxide production was increased and mitochondrial numbers decreased in the infected apoptotic subpopulation. Furthermore, a pan-caspase probe colocalised with SOD2 outside of mitochondria within cytosolic aggregates in infected cells and inhibition of caspase activity was able to restore cellular levels of SOD2 in the whole unseparated infected population to those of mock-infected control cells. Our results suggest that prion propagation exacerbates an apoptotic pathway whereby mitochondrial dysfunction follows mislocalisation of SOD2 to cytosolic caspases, permitting its degradation. Eventually, cellular capacity to maintain oxidative homeostasis is overwhelmed, thus resulting in cell death.
Collapse
Affiliation(s)
- Layla Sinclair
- Department of Pathology, Melbourne Brain Centre, The University of Melbourne, Victoria, 3010, Australia
| | | | | | | |
Collapse
|
40
|
Cellular aspects of prion replication in vitro. Viruses 2013; 5:374-405. [PMID: 23340381 PMCID: PMC3564126 DOI: 10.3390/v5010374] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 01/07/2013] [Accepted: 01/16/2013] [Indexed: 12/19/2022] Open
Abstract
Prion diseases or transmissible spongiform encephalopathies (TSEs) are fatal neurodegenerative disorders in mammals that are caused by unconventional agents predominantly composed of aggregated misfolded prion protein (PrP). Prions self-propagate by recruitment of host-encoded PrP into highly ordered β-sheet rich aggregates. Prion strains differ in their clinical, pathological and biochemical characteristics and are likely to be the consequence of distinct abnormal prion protein conformers that stably replicate their alternate states in the host cell. Understanding prion cell biology is fundamental for identifying potential drug targets for disease intervention. The development of permissive cell culture models has greatly enhanced our knowledge on entry, propagation and dissemination of TSE agents. However, despite extensive research, the precise mechanism of prion infection and potential strain effects remain enigmatic. This review summarizes our current knowledge of the cell biology and propagation of prions derived from cell culture experiments. We discuss recent findings on the trafficking of cellular and pathologic PrP, the potential sites of abnormal prion protein synthesis and potential co-factors involved in prion entry and propagation.
Collapse
|
41
|
Klemm HMJ, Welton JM, Masters CL, Klug GM, Boyd A, Hill AF, Collins SJ, Lawson VA. The prion protein preference of sporadic Creutzfeldt-Jakob disease subtypes. J Biol Chem 2012; 287:36465-72. [PMID: 22930754 DOI: 10.1074/jbc.m112.368803] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Sporadic Creutzfeldt-Jakob disease (CJD) is the most prevalent manifestation of the transmissible spongiform encephalopathies or prion diseases affecting humans. The disease encompasses a spectrum of clinical phenotypes that have been correlated with molecular subtypes that are characterized by the molecular mass of the protease-resistant fragment of the disease-related conformation of the prion protein and a polymorphism at codon 129 of the gene encoding the prion protein. A cell-free assay of prion protein misfolding was used to investigate the ability of these sporadic CJD molecular subtypes to propagate using brain-derived sources of the cellular prion protein (PrP(C)). This study confirmed the presence of three distinct sporadic CJD molecular subtypes with PrP(C) substrate requirements that reflected their codon 129 associations in vivo. However, the ability of a sporadic CJD molecular subtype to use a specific PrP(C) substrate was not determined solely by codon 129 as the efficiency of prion propagation was also influenced by the composition of the brain tissue from which the PrP(C) substrate was sourced, thus indicating that nuances in PrP(C) or additional factors may determine sporadic CJD subtype. The results of this study will aid in the design of diagnostic assays that can detect prion disease across the diversity of sporadic CJD subtypes.
Collapse
Affiliation(s)
- Helen M J Klemm
- Department of Pathology, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Coleman BM, Hanssen E, Lawson VA, Hill AF. Prion‐infected cells regulate the release of exosomes with distinct ultrastructural features. FASEB J 2012; 26:4160-73. [DOI: 10.1096/fj.11-202077] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Bradley M. Coleman
- Department of Biochemistry and Molecular BiologyThe University of MelbourneParkvilleVictoriaAustralia
- Department of PathologyThe University of MelbourneParkvilleVictoriaAustralia
- Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Eric Hanssen
- Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
- Bio21 Electron Microscopy UnitThe University of MelbourneParkvilleVictoriaAustralia
| | - Victoria A. Lawson
- Department of PathologyThe University of MelbourneParkvilleVictoriaAustralia
- The Mental Health Research InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Andrew F. Hill
- Department of Biochemistry and Molecular BiologyThe University of MelbourneParkvilleVictoriaAustralia
- Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
- The Mental Health Research InstituteThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
43
|
Prion subcellular fractionation reveals infectivity spectrum, with a high titre-low PrPres level disparity. Mol Neurodegener 2012; 7:18. [PMID: 22534096 PMCID: PMC3355018 DOI: 10.1186/1750-1326-7-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 04/26/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prion disease transmission and pathogenesis are linked to misfolded, typically protease resistant (PrPres) conformers of the normal cellular prion protein (PrPC), with the former posited to be the principal constituent of the infectious 'prion'. Unexplained discrepancies observed between detectable PrPres and infectivity levels exemplify the complexity in deciphering the exact biophysical nature of prions and those host cell factors, if any, which contribute to transmission efficiency. In order to improve our understanding of these important issues, this study utilized a bioassay validated cell culture model of prion infection to investigate discordance between PrPres levels and infectivity titres at a subcellular resolution. FINDINGS Subcellular fractions enriched in lipid rafts or endoplasmic reticulum/mitochondrial marker proteins were equally highly efficient at prion transmission, despite lipid raft fractions containing up to eight times the levels of detectable PrPres. Brain homogenate infectivity was not differentially enhanced by subcellular fraction-specific co-factors, and proteinase K pre-treatment of selected fractions modestly, but equally reduced infectivity. Only lipid raft associated infectivity was enhanced by sonication. CONCLUSIONS This study authenticates a subcellular disparity in PrPres and infectivity levels, and eliminates simultaneous divergence of prion strains as the explanation for this phenomenon. On balance, the results align best with the concept that transmission efficiency is influenced more by intrinsic characteristics of the infectious prion, rather than cellular microenvironment conditions or absolute PrPres levels.
Collapse
|
44
|
Gene knockout of tau expression does not contribute to the pathogenesis of prion disease. J Neuropathol Exp Neurol 2011; 70:1036-45. [PMID: 22002429 DOI: 10.1097/nen.0b013e318235b471] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Prion diseases or transmissible spongiform encephalopathies are a group of fatal and transmissible disorders affecting the central nervous system of humans and animals. The principal agent of prion disease transmission and pathogenesis is proposed to be an abnormal protease-resistant isoform of the normal cellular prion protein. The microtubule-associated protein tau is elevated in patients with Creutzfeldt-Jakob disease. To determine whether tau expression contributes to prion disease pathogenesis, tau knockout and control wild-type mice were infected with the M1000 strain of mouse-adapted human prions. Immunohistochemical analysis for total tau expression in prion-infected wild-type mice indicated tau aggregation in the cytoplasm of a subpopulation of neurons in regions associated with spongiform change. Western immunoblot analysis of brain homogenates revealed a decrease in total tau immunoreactivity and epitope-specific changes in tau phosphorylation. No significant difference in incubation period or other disease features were observed between tau knockout and wild-type mice with clinical prion disease. These results demonstrate that, in this model of prion disease, tau does not contribute to the pathogenesis of prion disease and that changes in the tau protein profile observed in mice with clinical prion disease occurs as a consequence of the prion-induced pathogenesis.
Collapse
|
45
|
Haigh CL, McGlade AR, Lewis V, Masters CL, Lawson VA, Collins SJ. Acute exposure to prion infection induces transient oxidative stress progressing to be cumulatively deleterious with chronic propagation in vitro. Free Radic Biol Med 2011; 51:594-608. [PMID: 21466851 DOI: 10.1016/j.freeradbiomed.2011.03.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Revised: 03/03/2011] [Accepted: 03/28/2011] [Indexed: 01/01/2023]
Abstract
Neuronal loss is a pathological feature of prion diseases for which increased reactive oxygen species (ROS) and consequent oxidative stress is one proposed mechanism. The processes underlying ROS production in prion disease and the precise relationship to misfolding of the prion protein remain obscure. Using cell culture models of prion infection we found that cells demonstrate a rapid, prion protein (PrP) dependent, increase in intracellular ROS following exposure to infectious inoculum. ROS production correlated with internalisation and increased intracellular protease resistant PrP (PrP(Res)). The ROS increase was predominantly lysosomal in origin but not sustained, with cells adapting within 48 hours. Overall ROS levels remained normal in the chronically prion infected cell population; however a subpopulation characterised by loss of membrane phosphatidylserine asymmetry exhibited highly peroxidised intracellular aggregates that localised with PrP and intense caspase activation. These apoptotic cells showed increased ROS closely correlating with increased PrP(Res). Our findings demonstrate that a PrP-dependent, transient, increase in intracellular ROS is characteristic of acute cellular prion infection, while chronic phases of prion infection in vitro are associated with a significant subpopulation manifesting apoptosis accompanying heightened oxidative stress and increased PrP(Res) burden. Such observations strengthen the direct links between heightened ROS and ongoing prion propagation with eventual cellular demise.
Collapse
Affiliation(s)
- Cathryn L Haigh
- Department of Pathology, The University of Melbourne, Parkville, 3010, Australia
| | | | | | | | | | | |
Collapse
|
46
|
A simple, versatile and sensitive cell-based assay for prions from various species. PLoS One 2011; 6:e20563. [PMID: 21655184 PMCID: PMC3105100 DOI: 10.1371/journal.pone.0020563] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 05/03/2011] [Indexed: 12/03/2022] Open
Abstract
Detection and quantification of prion infectivity is a crucial step for various fundamental and applied aspects of prion research. Identification of cell lines highly sensitive to prion infection led to the development of cell-based titration procedures aiming at replacing animal bioassays, usually performed in mice or hamsters. However, most of these cell lines are only permissive to mouse-adapted prions strains and do not allow titration of prions from other species. In this study, we show that epithelial RK13, a cell line permissive to mouse and bank vole prion strains and to natural prion agents from sheep and cervids, enables a robust and sensitive detection of mouse and ovine-derived prions. Importantly, the cell culture work is strongly reduced as the RK13 cell assay procedure designed here does not require subcultivation of the inoculated cultures. We also show that prions effectively bind to culture plastic vessel and are quantitatively detected by the cell assay. The possibility to easily quantify a wider range of prions, including rodent experimental strains but also natural agents from sheep and cervids, should prompt the spread of cell assays for routine prion titration and lead to valuable information in fundamental and applied studies.
Collapse
|
47
|
Lawson VA, Haigh CL, Roberts B, Kenche VB, Klemm HMJ, Masters CL, Collins SJ, Barnham KJ, Drew SC. Near-infrared fluorescence imaging of apoptotic neuronal cell death in a live animal model of prion disease. ACS Chem Neurosci 2010; 1:720-7. [PMID: 22778809 DOI: 10.1021/cn100068x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 09/17/2010] [Indexed: 11/29/2022] Open
Abstract
Apoptotic cell death via activation of the caspase family of cysteine proteases is a common feature of many neurodegenerative diseases including Creutzfeldt-Jakob disease. Molecular imaging of cysteine protease activities at the preclinical stage may provide valuable mechanistic information about pathophysiological pathways involved in disease evolution and in response to therapy. In this study, we report synthesis and characterization of a near-infrared (NIR) fluorescent contrast agent capable of noninvasively imaging neuronal apoptosis in vivo, by conjugating a NIR cyanine dye to Val-Ala-Asp-fluoromethylketone (VAD-fmk), a general inhibitor of active caspases. Following intravenous administration of the NIR-VAD-fmk contrast agent, in vivo fluorescence reflectance imaging identified significantly higher levels of active caspases in the brain of mice with advanced but preclinical prion disease, when compared with healthy controls. The contrast agent and related analogues will enable the longitudinal study of disease progression and therapy in animal models of many neurodegenerative conditions.
Collapse
Affiliation(s)
- Victoria A. Lawson
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
- Mental Health Research Institute, Parkville, Victoria 3052, Australia
| | - Cathryn L. Haigh
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
- Mental Health Research Institute, Parkville, Victoria 3052, Australia
| | - Blaine Roberts
- Mental Health Research Institute, Parkville, Victoria 3052, Australia
- Centre for Neuroscience, The University of Melbourne, Victoria 3010, Australia
| | - Vijaya B. Kenche
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
- Mental Health Research Institute, Parkville, Victoria 3052, Australia
- The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Helen M. J. Klemm
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
| | - Colin L. Masters
- Mental Health Research Institute, Parkville, Victoria 3052, Australia
- Centre for Neuroscience, The University of Melbourne, Victoria 3010, Australia
| | - Steven J. Collins
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
- Mental Health Research Institute, Parkville, Victoria 3052, Australia
| | - Kevin J. Barnham
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
- Mental Health Research Institute, Parkville, Victoria 3052, Australia
- The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Simon C. Drew
- Department of Pathology, The University of Melbourne, Victoria 3010, Australia
- Mental Health Research Institute, Parkville, Victoria 3052, Australia
- The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
48
|
Lawson VA, Lumicisi B, Welton J, Machalek D, Gouramanis K, Klemm HM, Stewart JD, Masters CL, Hoke DE, Collins SJ, Hill AF. Glycosaminoglycan sulphation affects the seeded misfolding of a mutant prion protein. PLoS One 2010; 5:e12351. [PMID: 20808809 PMCID: PMC2925953 DOI: 10.1371/journal.pone.0012351] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 07/22/2010] [Indexed: 11/18/2022] Open
Abstract
Background The accumulation of protease resistant conformers of the prion protein (PrPres) is a key pathological feature of prion diseases. Polyanions, including RNA and glycosaminoglycans have been identified as factors that contribute to the propagation, transmission and pathogenesis of prion disease. Recent studies have suggested that the contribution of these cofactors to prion propagation may be species specific. Methodology/Principal Finding In this study a cell-free assay was used to investigate the molecular basis of polyanion stimulated PrPres formation using brain tissue or cell line derived murine PrP. Enzymatic depletion of endogenous nucleic acids or heparan sulphate (HS) from the PrPC substrate was found to specifically prevent PrPres formation seeded by mouse derived PrPSc. Modification of the negative charge afforded by the sulphation of glycosaminoglycans increased the ability of a familial PrP mutant to act as a substrate for PrPres formation, while having no effect on PrPres formed by wildtype PrP. This difference may be due to the observed differences in the binding of wild type and mutant PrP for glycosaminoglycans. Conclusions/Significance Cofactor requirements for PrPres formation are host species and prion strain specific and affected by disease associated mutations of the prion protein. This may explain both species and strain dependent propagation characteristics and provide insights into the underlying mechanisms of familial prion disease. It further highlights the challenge of designing effective therapeutics against a disease which effects a range of mammalian species, caused by range of aetiologies and prion strains.
Collapse
Affiliation(s)
- Victoria A. Lawson
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
- The Mental Health Research Institute, The University of Melbourne, Parkville, Victoria, Australia
- * E-mail: (VAL); (AFH)
| | - Brooke Lumicisi
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jeremy Welton
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Dorothy Machalek
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Katrina Gouramanis
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Helen M. Klemm
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - James D. Stewart
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L. Masters
- The Mental Health Research Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - David E. Hoke
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Steven J. Collins
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
- The Mental Health Research Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Andrew F. Hill
- Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry & Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- The Mental Health Research Institute, The University of Melbourne, Parkville, Victoria, Australia
- * E-mail: (VAL); (AFH)
| |
Collapse
|
49
|
Abstract
Cell-based measurement of prion infectivity is currently restricted to experimental strains of mouse-adapted scrapie. Having isolated cell cultures with susceptibility to prions from diseased elk, we describe a modification of the scrapie cell assay allowing evaluation of prions causing chronic wasting disease, a naturally occurring transmissible spongiform encephalopathy. We compare this cervid prion cell assay to bioassays in transgenic mice, the only other existing method for quantification, and show this assay to be a relatively economical and expedient alternative that will likely facilitate studies of this important prion disease.
Collapse
|
50
|
Changing the solvent accessibility of the prion protein disulfide bond markedly influences its trafficking and effect on cell function. Biochem J 2010; 428:169-82. [PMID: 20337594 DOI: 10.1042/bj20091635] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Prion diseases are fatal transmissible neurodegenerative diseases that result from structural conversion of the prion protein into a disease-associated isoform. The prion protein contains a single disulfide bond. Our analysis of all NMR structures of the prion protein (total of 440 structures over nine species) containing an explicit disulfide bond reveals that the bond exists predominantly in a stable low-energy state, but can also adopt a high-energy configuration. The side chains of two tyrosine residues and one phenylalanine residue control access of solvent to the disulfide bond. Notably, the side chains rotate away from the disulfide bond in the high-energy state, exposing the disulfide bond to solvent. The importance of these aromatic residues for protein function was analysed by mutating them to alanine residues and analysing the properties of the mutant proteins using biophysical and cell biological approaches. Whereas the mutant protein behaved similarly to wild-type prion protein in recombinant systems, the mutants were retained in the endoplasmic reticulum of mammalian cells and degraded by the proteasomal system. The cellular behaviour of the aromatic residue mutants was similar to the cellular behaviour of a disulfide bond mutant prion protein in which the cysteine residues were replaced with alanine, a result which is consistent with an unstable disulfide bond in the aromatic residue mutants. These observations suggest that the conformation of the prion protein disulfide bond may have implications for correct maturation and function of this protein.
Collapse
|