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; 102:448-471. [PMID: 38996387 DOI: 10.1139/bcb-2024-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 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
|
Gojanovich AD, Le NTT, Mercer RCC, Park S, Wu B, Anane A, Vultaggio JS, Mostoslavsky G, Harris DA. Abnormal synaptic architecture in iPSC-derived neurons from a multi-generational family with genetic Creutzfeldt-Jakob disease. Stem Cell Reports 2024; 19:1474-1488. [PMID: 39332406 PMCID: PMC11561462 DOI: 10.1016/j.stemcr.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/29/2024] Open
Abstract
Genetic prion diseases are caused by mutations in PRNP, which encodes the prion protein (PrPC). Why these mutations are pathogenic, and how they alter the properties of PrPC are poorly understood. We have consented and accessed 22 individuals of a multi-generational Israeli family harboring the highly penetrant E200K PRNP mutation and generated a library of induced pluripotent stem cells (iPSCs) representing nine carriers and four non-carriers. iPSC-derived neurons from E200K carriers display abnormal synaptic architecture characterized by misalignment of postsynaptic NMDA receptors with the cytoplasmic scaffolding protein PSD95. Differentiated neurons from mutation carriers do not produce PrPSc, the aggregated and infectious conformer of PrP, suggesting that loss of a physiological function of PrPC may contribute to the disease phenotype. Our study shows that iPSC-derived neurons can provide important mechanistic insights into the pathogenesis of genetic prion diseases and can offer a powerful platform for testing candidate therapeutics.
Collapse
Affiliation(s)
- Aldana D Gojanovich
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
| | - Nhat T T Le
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Robert C C Mercer
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Seonmi Park
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
| | - Bei Wu
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Alice Anane
- Creutzfeldt-Jakob Disease Foundation, Pardes Hanna-Karkur, Israel
| | - Janelle S Vultaggio
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Gustavo Mostoslavsky
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA; Department of Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - David A Harris
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
3
|
Mehra S, Bourkas ME, Kaczmarczyk L, Stuart E, Arshad H, Griffin JK, Frost KL, Walsh DJ, Supattapone S, Booth SA, Jackson WS, Watts JC. Convergent generation of atypical prions in knockin mouse models of genetic prion disease. J Clin Invest 2024; 134:e176344. [PMID: 39087478 PMCID: PMC11291267 DOI: 10.1172/jci176344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 06/11/2024] [Indexed: 08/02/2024] Open
Abstract
Most cases of human prion disease arise due to spontaneous misfolding of WT or mutant prion protein, yet recapitulating this event in animal models has proven challenging. It remains unclear whether spontaneous prion generation can occur within the mouse lifespan in the absence of protein overexpression and how disease-causing mutations affect prion strain properties. To address these issues, we generated knockin mice that express the misfolding-prone bank vole prion protein (BVPrP). While mice expressing WT BVPrP (I109 variant) remained free from neurological disease, a subset of mice expressing BVPrP with mutations (D178N or E200K) causing genetic prion disease developed progressive neurological illness. Brains from spontaneously ill knockin mice contained prion disease-specific neuropathological changes as well as atypical protease-resistant BVPrP. Moreover, brain extracts from spontaneously ill D178N- or E200K-mutant BVPrP-knockin mice exhibited prion seeding activity and transmitted disease to mice expressing WT BVPrP. Surprisingly, the properties of the D178N- and E200K-mutant prions appeared identical before and after transmission, suggesting that both mutations guide the formation of a similar atypical prion strain. These findings imply that knockin mice expressing mutant BVPrP spontaneously develop a bona fide prion disease and that mutations causing prion diseases may share a uniform initial mechanism of action.
Collapse
Affiliation(s)
- Surabhi Mehra
- Tanz Centre for Research in Neurodegenerative Diseases and
| | - Matthew E.C. Bourkas
- Tanz Centre for Research in Neurodegenerative Diseases and
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Lech Kaczmarczyk
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Erica Stuart
- Tanz Centre for Research in Neurodegenerative Diseases and
| | - Hamza Arshad
- Tanz Centre for Research in Neurodegenerative Diseases and
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | | | - Kathy L. Frost
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | | | - Surachai Supattapone
- Department of Biochemistry and Cell Biology and
- Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Stephanie A. Booth
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Walker S. Jackson
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Joel C. Watts
- Tanz Centre for Research in Neurodegenerative Diseases and
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Walsh DJ, Rees JR, Mehra S, Bourkas MEC, Kaczmarczyk L, Stuart E, Jackson WS, Watts JC, Supattapone S. Anti-prion drugs do not improve survival in novel knock-in models of inherited prion disease. PLoS Pathog 2024; 20:e1012087. [PMID: 38557815 PMCID: PMC10984475 DOI: 10.1371/journal.ppat.1012087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Prion diseases uniquely manifest in three distinct forms: inherited, sporadic, and infectious. Wild-type prions are responsible for the sporadic and infectious versions, while mutant prions cause inherited variants like fatal familial insomnia (FFI) and familial Creutzfeldt-Jakob disease (fCJD). Although some drugs can prolong prion incubation times up to four-fold in rodent models of infectious prion diseases, no effective treatments for FFI and fCJD have been found. In this study, we evaluated the efficacy of various anti-prion drugs on newly-developed knock-in mouse models for FFI and fCJD. These models express bank vole prion protein (PrP) with the pathogenic D178N and E200K mutations. We applied various drug regimens known to be highly effective against wild-type prions in vivo as well as a brain-penetrant compound that inhibits mutant PrPSc propagation in vitro. None of the regimens tested (Anle138b, IND24, Anle138b + IND24, cellulose ether, and PSCMA) significantly extended disease-free survival or prevented mutant PrPSc accumulation in either knock-in mouse model, despite their ability to induce strain adaptation of mutant prions. Our results show that anti-prion drugs originally developed to treat infectious prion diseases do not necessarily work for inherited prion diseases, and that the recombinant sPMCA is not a reliable platform for identifying compounds that target mutant prions. This work underscores the need to develop therapies and validate screening assays specifically for mutant prions, as well as anti-prion strategies that are not strain-dependent.
Collapse
Affiliation(s)
- Daniel J. Walsh
- Department of Biochemistry and Cell Biology Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Judy R. Rees
- Department of Epidemiology Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- Department of Community and Family Medicine Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Surabhi Mehra
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Matthew E. C. Bourkas
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Lech Kaczmarczyk
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Erica Stuart
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Walker S. Jackson
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Joel C. Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Surachai Supattapone
- Department of Biochemistry and Cell Biology Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|
5
|
Jackson WS, Bauer S, Kaczmarczyk L, Magadi SS. Selective Vulnerability to Neurodegenerative Disease: Insights from Cell Type-Specific Translatome Studies. BIOLOGY 2024; 13:67. [PMID: 38392286 PMCID: PMC10886597 DOI: 10.3390/biology13020067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/24/2024]
Abstract
Neurodegenerative diseases (NDs) manifest a wide variety of clinical symptoms depending on the affected brain regions. Gaining insights into why certain regions are resistant while others are susceptible is vital for advancing therapeutic strategies. While gene expression changes offer clues about disease responses across brain regions, the mixture of cell types therein obscures experimental results. In recent years, methods that analyze the transcriptomes of individual cells (e.g., single-cell RNA sequencing or scRNAseq) have been widely used and have provided invaluable insights into specific cell types. Concurrently, transgene-based techniques that dissect cell type-specific translatomes (CSTs) in model systems, like RiboTag and bacTRAP, offer unique advantages but have received less attention. This review juxtaposes the merits and drawbacks of both methodologies, focusing on the use of CSTs in understanding conditions like amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), Alzheimer's disease (AD), and specific prion diseases like fatal familial insomnia (FFI), genetic Creutzfeldt-Jakob disease (gCJD), and acquired prion disease. We conclude by discussing the emerging trends observed across multiple diseases and emerging methods.
Collapse
Affiliation(s)
- Walker S Jackson
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Susanne Bauer
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Lech Kaczmarczyk
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Srivathsa S Magadi
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| |
Collapse
|
6
|
Jackson WS. Etiology matters: genetic and acquired prion diseases engage different mechanisms at a presymptomatic stage. Neural Regen Res 2023; 18:2707-2708. [PMID: 37449633 DOI: 10.4103/1673-5374.373684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Affiliation(s)
- Walker S Jackson
- Department of Biomedical and Clinical Sciences, Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
7
|
Walsh DJ, Rees JR, Mehra S, Bourkas MEC, Kaczmarczyk L, Stuart E, Jackson WS, Watts JC, Supattapone S. Anti-prion drugs do not improve survival in knock-in models of inherited prion disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.559951. [PMID: 37808761 PMCID: PMC10557747 DOI: 10.1101/2023.09.28.559951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Prion diseases uniquely manifest in three distinct forms: inherited, sporadic, and infectious. Wild-type prions are responsible for the sporadic and infectious versions, while mutant prions cause inherited variants like fatal familial insomnia (FFI) and familial Creutzfeldt-Jakob disease (fCJD). Although some drugs can prolong prion incubation times up to four-fold in rodent models of infectious prion diseases, no effective treatments for FFI and fCJD have been found. In this study, we evaluated the efficacy of various anti-prion drugs on newly-developed knock-in mouse models for FFI and fCJD. These models express bank vole prion protein (PrP) with the pathogenic D178N and E200K mutations. We applied various drug regimens known to be highly effective against wild-type prions in vivo as well as a brain-penetrant compound that inhibits mutant PrP Sc propagation in vitro . None of the regimens tested (Anle138b, IND24, Anle138b + IND24, cellulose ether, and PSCMA) significantly extended disease-free survival or prevented mutant PrP Sc accumulation in either knock-in mouse model, despite their ability to induce strain adaptation of mutant prions. Paradoxically, the combination of Anle138b and IND24 appeared to accelerate disease by 16% and 26% in kiBVI E200K and kiBVI D178N mice, respectively, and accelerated the aggregation of mutant PrP molecules in vitro . Our results show that anti-prion drugs originally developed to treat infectious prion diseases do not necessarily work for inherited prion diseases, and that the recombinant sPMCA is not a reliable platform for identifying compounds that target mutant prions. This work underscores the need to develop therapies and validate screening assays specifically for mutant prions.
Collapse
|
8
|
Bordonaro M. Hypothesis: functional age and onset of autosomal dominant genetic prion disease. Theory Biosci 2023; 142:143-150. [PMID: 37017882 DOI: 10.1007/s12064-023-00389-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 03/10/2023] [Indexed: 04/06/2023]
Abstract
Autosomal dominant diseases typically have an age-related onset. Here, I focus on genetic prion disease (gPrD), caused by various mutations in the PRNP gene. While gPrD typically occurs at or after middle age, there can be considerable variability in the specific age of onset. This variability can occur among patients with the same PRNP mutation; in some cases, these differences occur not only between families but even within the same family. It is not known why gPrD onset is typically delayed for decades when the causative mutation is present from birth. Mouse models of gPrD manifest disease; however, unlike human gPrD, which typically takes decades to manifest, mouse models exhibit disease within months. Therefore, the time to onset of prion disease is proportional to species lifespan; however, it is not known why this is the case. I hypothesize that the initiation of gPrD is strongly influenced by the process of aging; therefore, disease onset is related to proportional functional age (e.g., mice vs. humans). I propose approaches to test this hypothesis and discuss its significance with respect to delaying prion disease through suppression of aging.
Collapse
Affiliation(s)
- Michael Bordonaro
- Department of Medical Education, Geisinger Commonwealth School of Medicine, 525 Pine Street, Scranton, PA, 18509, USA.
| |
Collapse
|
9
|
Vallabh SM, Zou D, Pitstick R, O’Moore J, Peters J, Silvius D, Kriz J, Jackson WS, Carlson GA, Minikel EV, Cabin DE. Therapeutic Trial of anle138b in Mouse Models of Genetic Prion Disease. J Virol 2023; 97:e0167222. [PMID: 36651748 PMCID: PMC9973041 DOI: 10.1128/jvi.01672-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Phenotypic screening has yielded small-molecule inhibitors of prion replication that are effective in vivo against certain prion strains but not others. Here, we sought to test the small molecule anle138b in multiple mouse models of prion disease. In mice inoculated with the RML strain of prions, anle138b doubled survival and durably suppressed astrogliosis measured by live-animal bioluminescence imaging. In knock-in mouse models of the D178N and E200K mutations that cause genetic prion disease, however, we were unable to identify a clear, quantifiable disease endpoint against which to measure therapeutic efficacy. Among untreated animals, the mutations did not impact overall survival, and bioluminescence remained low out to >20 months of age. Vacuolization and PrP deposition were observed in some brain regions in a subset of mutant animals but appeared to be unable to carry the weight of a primary endpoint in a therapeutic study. We conclude that not all animal models of prion disease are suited to well-powered therapeutic efficacy studies, and care should be taken in choosing the models that will support drug development programs. IMPORTANCE There is an urgent need to develop drugs for prion disease, a currently untreatable neurodegenerative disease. In this effort, there is a debate over which animal models can best support a drug development program. While the study of prion disease benefits from excellent animal models because prions naturally afflict many different mammals, different models have different capabilities and limitations. Here, we conducted a therapeutic efficacy study of the drug candidate anle138b in mouse models with two of the most common mutations that cause genetic prion disease. In a more typical model where prions are injected directly into the brain, we found anle138b to be effective. In the genetic models, however, the animals never reached a clear, measurable point of disease onset. We conclude that not all prion disease animal models are ideally suited to drug efficacy studies, and well-defined, quantitative disease metrics should be a priority.
Collapse
Affiliation(s)
- Sonia M. Vallabh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
- Prion Alliance, Cambridge, Massachusetts, USA
| | - Dan Zou
- Montana Veterinary Diagnostic Laboratory, Bozeman, Montana, USA
| | - Rose Pitstick
- McLaughlin Research Institute, Great Falls, Montana, USA
| | - Jill O’Moore
- McLaughlin Research Institute, Great Falls, Montana, USA
| | - Janet Peters
- McLaughlin Research Institute, Great Falls, Montana, USA
| | - Derek Silvius
- McLaughlin Research Institute, Great Falls, Montana, USA
| | - Jasna Kriz
- Cervo Brain Research Center, Université Laval, Québec, Québec, Canada
| | - Walker S. Jackson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - George A. Carlson
- Institute for Neurodegenerative Diseases, University of California—San Francisco, San Francisco, California, USA
| | - Eric Vallabh Minikel
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
- Prion Alliance, Cambridge, Massachusetts, USA
| | | |
Collapse
|
10
|
Bauer S, Dittrich L, Kaczmarczyk L, Schleif M, Benfeitas R, Jackson WS. Translatome profiling in fatal familial insomnia implicates TOR signaling in somatostatin neurons. Life Sci Alliance 2022; 5:5/11/e202201530. [PMID: 36192034 PMCID: PMC9531780 DOI: 10.26508/lsa.202201530] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/24/2022] Open
Abstract
Bauer and colleagues report that among the six neuron types studied, somatostatin neurons have an unexpectedly strong and similar response to two distinct genetic prion diseases before disease onset. Selective neuronal vulnerability is common in neurodegenerative diseases but poorly understood. In genetic prion diseases, including fatal familial insomnia (FFI) and Creutzfeldt–Jakob disease (CJD), different mutations in the Prnp gene manifest as clinically and neuropathologically distinct diseases. Here we report with electroencephalography studies that theta waves are mildly increased in 21 mo old knock-in mice modeling FFI and CJD and that sleep is mildy affected in FFI mice. To define affected cell types, we analyzed cell type–specific translatomes from six neuron types of 9 mo old FFI and CJD mice. Somatostatin (SST) neurons responded the strongest in both diseases, with unexpectedly high overlap in genes and pathways. Functional analyses revealed up-regulation of neurodegenerative disease pathways and ribosome and mitochondria biogenesis, and down-regulation of synaptic function and small GTPase-mediated signaling in FFI, implicating down-regulation of mTOR signaling as the root of these changes. In contrast, responses in glutamatergic cerebellar neurons were disease-specific. The high similarity in SST neurons of FFI and CJD mice suggests that a common therapy may be beneficial for multiple genetic prion diseases.
Collapse
Affiliation(s)
- Susanne Bauer
- Department of Biomedical and Clinical Sciences, Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Lars Dittrich
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Lech Kaczmarczyk
- Department of Biomedical and Clinical Sciences, Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden.,German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Melvin Schleif
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Rui Benfeitas
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Walker S Jackson
- Department of Biomedical and Clinical Sciences, Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden .,German Center for Neurodegenerative Diseases, Bonn, Germany
| |
Collapse
|
11
|
Kaczmarczyk L, Schleif M, Dittrich L, Williams RH, Koderman M, Bansal V, Rajput A, Schulte T, Jonson M, Krost C, Testaquadra FJ, Bonn S, Jackson WS. Distinct translatome changes in specific neural populations precede electroencephalographic changes in prion-infected mice. PLoS Pathog 2022; 18:e1010747. [PMID: 35960762 PMCID: PMC9401167 DOI: 10.1371/journal.ppat.1010747] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 08/24/2022] [Accepted: 07/18/2022] [Indexed: 12/04/2022] Open
Abstract
Selective vulnerability is an enigmatic feature of neurodegenerative diseases (NDs), whereby a widely expressed protein causes lesions in specific cell types and brain regions. Using the RiboTag method in mice, translational responses of five neural subtypes to acquired prion disease (PrD) were measured. Pre-onset and disease onset timepoints were chosen based on longitudinal electroencephalography (EEG) that revealed a gradual increase in theta power between 10- and 18-weeks after prion injection, resembling a clinical feature of human PrD. At disease onset, marked by significantly increased theta power and histopathological lesions, mice had pronounced translatome changes in all five cell types despite appearing normal. Remarkably, at a pre-onset stage, prior to EEG and neuropathological changes, we found that 1) translatomes of astrocytes indicated reduced synthesis of ribosomal and mitochondrial components, 2) glutamatergic neurons showed increased expression of cytoskeletal genes, and 3) GABAergic neurons revealed reduced expression of circadian rhythm genes. These data demonstrate that early translatome responses to neurodegeneration emerge prior to conventional markers of disease and are cell type-specific. Therapeutic strategies may need to target multiple pathways in specific populations of cells, early in disease. Prions are infectious agents composed of a misfolded protein. When isolated from a mammalian brain and transferred to the same host species, prions will cause the same neurodegenerative disease affecting the same brain regions and cell types. This concept of selective vulnerability is also a feature of more common types of neurodegenerative diseases, such as Alzheimer’s, Parkinson’s, and Huntington’s. To better understand the mechanisms behind selective vulnerability, we studied disease responses of five cell types with different vulnerabilities in prion-infected mice at two different disease stages. Responses were measured as changes to mRNAs undergoing translation, referred to as the translatome. Before prion-infected mice demonstrated typical disease signs, electroencephalography (a method used clinically to characterize neurodegeneration in humans) revealed brain changes resembling those in human prion diseases, and surprisingly, the translatomes of all cells were drastically changed. Furthermore, before electroencephalography changes emerged, three cell types made unique responses while the most vulnerable cell type did not. These results suggests that mechanisms causing selective vulnerability will be difficult to dissect and that therapies will likely need to be provided before clinical signs emerge and individually engage multiple cell types and their distinct molecular pathways.
Collapse
Affiliation(s)
- Lech Kaczmarczyk
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Melvin Schleif
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Lars Dittrich
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | - Maruša Koderman
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Vikas Bansal
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Ashish Rajput
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Germany
- Maximon AG, Zug, Switzerland
| | | | - Maria Jonson
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Clemens Krost
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | - Stefan Bonn
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Germany
| | - Walker S. Jackson
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- German Center for Neurodegenerative Diseases, Bonn, Germany
- * E-mail:
| |
Collapse
|
12
|
Luan X, Zhang X, Zhou Y. The Role and Clinical Observation of Traditional Chinese Medicine in Relieving Senile Insomnia: A Systematic Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9484095. [PMID: 35601141 PMCID: PMC9122729 DOI: 10.1155/2022/9484095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/22/2022] [Accepted: 03/26/2022] [Indexed: 12/16/2022]
Abstract
Objective To assess the role and clinical observation of traditional Chinese medicine (TCM) in relieving senile insomnia and conduct a systematic evaluation and meta-analysis. Methods We searched Chinese literatures from some database, including the China Knowledge Network database, Chongqing CQVIP Chinese database, Wanfang database, and Chinese Biomedical Literature database. In addition, English literature retrieval database PubMed and the Cochrane Library were included in this meta-analysis. The randomized controlled trial (RCT) of cognitive behavioral therapy was found in the treatment of chronic insomnia. All articles were current up to December 1, 2021, and references in the literature were hand searched. Results About 16 studies were included, involving 1260 elderly patients with insomnia. Meta-analysis results showed that compared with the control group, the sleep latency of the experimental group MD = -8.02, 95% CI (10.95, 5.01), the number of awakenings of the experimental group MD = -0.41, 95% CI (0.68, 0.14), the total sleep time of the experimental group MD = 39.41, 95% CI (27.05, 51.71), the sleep efficiency of the experimental group MD = 7.82, 95% CI (1.87, 13.75) the Pittsburgh sleep quality index of the experimental group MD = 2.41, 95% CI (3.12, 1.46), the effective rate of the experimental group: OR = 3.12, 95% CI (1.76, 5.48), and the insomnia severity index of the experimental group MD = 1.05, 95% CI (2.04, 0.52). Conclusion Our current study has indicated that TCM may play a central role in relieving senile insomnia and may be effective when treating insomnia. However, due to the low quality of trial methodology and publication bias, it cannot be fully confirmed. Further high-quality trials are needed to be investigated in the future.
Collapse
Affiliation(s)
- Xin Luan
- Department of Traditional Medicine, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
| | - Xiaodan Zhang
- Department of Traditional Medicine, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
| | - Yixin Zhou
- Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine 200120, Shanghai 200120, China
| |
Collapse
|
13
|
Virus Infection, Genetic Mutations, and Prion Infection in Prion Protein Conversion. Int J Mol Sci 2021; 22:ijms222212439. [PMID: 34830321 PMCID: PMC8624980 DOI: 10.3390/ijms222212439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 01/04/2023] Open
Abstract
Conformational conversion of the cellular isoform of prion protein, PrPC, into the abnormally folded, amyloidogenic isoform, PrPSc, is an underlying pathogenic mechanism in prion diseases. The diseases manifest as sporadic, hereditary, and acquired disorders. Etiological mechanisms driving the conversion of PrPC into PrPSc are unknown in sporadic prion diseases, while prion infection and specific mutations in the PrP gene are known to cause the conversion of PrPC into PrPSc in acquired and hereditary prion diseases, respectively. We recently reported that a neurotropic strain of influenza A virus (IAV) induced the conversion of PrPC into PrPSc as well as formation of infectious prions in mouse neuroblastoma cells after infection, suggesting the causative role of the neuronal infection of IAV in sporadic prion diseases. Here, we discuss the conversion mechanism of PrPC into PrPSc in different types of prion diseases, by presenting our findings of the IAV infection-induced conversion of PrPC into PrPSc and by reviewing the so far reported transgenic animal models of hereditary prion diseases and the reverse genetic studies, which have revealed the structure-function relationship for PrPC to convert into PrPSc after prion infection.
Collapse
|
14
|
Slc1a3-2A-CreERT2 mice reveal unique features of Bergmann glia and augment a growing collection of Cre drivers and effectors in the 129S4 genetic background. Sci Rep 2021; 11:5412. [PMID: 33686166 PMCID: PMC7940647 DOI: 10.1038/s41598-021-84887-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 02/22/2021] [Indexed: 12/26/2022] Open
Abstract
Genetic variation is a primary determinant of phenotypic diversity. In laboratory mice, genetic variation can be a serious experimental confounder, and thus minimized through inbreeding. However, generalizations of results obtained with inbred strains must be made with caution, especially when working with complex phenotypes and disease models. Here we compared behavioral characteristics of C57Bl/6—the strain most widely used in biomedical research—with those of 129S4. In contrast to 129S4, C57Bl/6 demonstrated high within-strain and intra-litter behavioral hyperactivity. Although high consistency would be advantageous, the majority of disease models and transgenic tools are in C57Bl/6. We recently established six Cre driver lines and two Cre effector lines in 129S4. To augment this collection, we genetically engineered a Cre line to study astrocytes in 129S4. It was validated with two Cre effector lines: calcium indicator gCaMP5g-tdTomato and RiboTag—a tool widely used to study cell type-specific translatomes. These reporters are in different genomic loci, and in both the Cre was functional and astrocyte-specific. We found that calcium signals lasted longer and had a higher amplitude in cortical compared to hippocampal astrocytes, genes linked to a single neurodegenerative disease have highly divergent expression patterns, and that ribosome proteins are non-uniformly expressed across brain regions and cell types.
Collapse
|
15
|
Marín-Moreno A, Espinosa JC, Torres JM. Transgenic mouse models for the study of prion diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:147-177. [PMID: 32958231 DOI: 10.1016/bs.pmbts.2020.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Prions are unique agents that challenge the molecular biology dogma by transmitting information on the protein level. They cause neurodegenerative diseases that lack of any cure or treatment called transmissible spongiform encephalopathies. The function of the normal form of the prion protein, the exact mechanism of prion propagation between species as well as at the cellular level and neuron degeneration remains elusive. However, great amount of information known for all these aspects has been achieved thanks to the use of animal models and more precisely to transgenic mouse models. In this chapter, the main contributions of these powerful research tools in the prion field are revised.
Collapse
Affiliation(s)
- Alba Marín-Moreno
- Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Spain
| | | | - Juan María Torres
- Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Spain.
| |
Collapse
|
16
|
Munoz-Montesino C, Larkem D, Barbereau C, Igel-Egalon A, Truchet S, Jacquet E, Nhiri N, Moudjou M, Sizun C, Rezaei H, Béringue V, Dron M. A seven-residue deletion in PrP leads to generation of a spontaneous prion formed from C-terminal C1 fragment of PrP. J Biol Chem 2020; 295:14025-14039. [PMID: 32788216 DOI: 10.1074/jbc.ra120.014738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Prions result from a drastic conformational change of the host-encoded cellular prion protein (PrP), leading to the formation of β-sheet-rich, insoluble, and protease-resistant self-replicating assemblies (PrPSc). The cellular and molecular mechanisms involved in spontaneous prion formation in sporadic and inherited human prion diseases or equivalent animal diseases are poorly understood, in part because cell models of spontaneously forming prions are currently lacking. Here, extending studies on the role of the H2 α-helix C terminus of PrP, we found that deletion of the highly conserved 190HTVTTTT196 segment of ovine PrP led to spontaneous prion formation in the RK13 rabbit kidney cell model. On long-term passage, the mutant cells stably produced proteinase K (PK)-resistant, insoluble, and aggregated assemblies that were infectious for naïve cells expressing either the mutant protein or other PrPs with slightly different deletions in the same area. The electrophoretic pattern of the PK-resistant core of the spontaneous prion (ΔSpont) contained mainly C-terminal polypeptides akin to C1, the cell-surface anchored C-terminal moiety of PrP generated by natural cellular processing. RK13 cells expressing solely the Δ190-196 C1 PrP construct, in the absence of the full-length protein, were susceptible to ΔSpont prions. ΔSpont infection induced the conversion of the mutated C1 into a PK-resistant and infectious form perpetuating the biochemical characteristics of ΔSpont prion. In conclusion, this work provides a unique cell-derived system generating spontaneous prions and provides evidence that the 113 C-terminal residues of PrP are sufficient for a self-propagating prion entity.
Collapse
Affiliation(s)
- Carola Munoz-Montesino
- Université Paris-Saclay, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université de Versailles Saint-Quentin-en-Yvelines, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Djabir Larkem
- Université Paris-Saclay, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université de Versailles Saint-Quentin-en-Yvelines, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Clément Barbereau
- Université Paris-Saclay, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université de Versailles Saint-Quentin-en-Yvelines, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Angélique Igel-Egalon
- Université Paris-Saclay, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université de Versailles Saint-Quentin-en-Yvelines, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Sandrine Truchet
- Université Paris-Saclay, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université de Versailles Saint-Quentin-en-Yvelines, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Eric Jacquet
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Naïma Nhiri
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Mohammed Moudjou
- Université Paris-Saclay, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université de Versailles Saint-Quentin-en-Yvelines, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Christina Sizun
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Human Rezaei
- Université Paris-Saclay, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université de Versailles Saint-Quentin-en-Yvelines, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Vincent Béringue
- Université Paris-Saclay, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université de Versailles Saint-Quentin-en-Yvelines, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Michel Dron
- Université Paris-Saclay, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université de Versailles Saint-Quentin-en-Yvelines, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| |
Collapse
|
17
|
Transcriptional signature of prion-induced neurotoxicity in a Drosophila model of transmissible mammalian prion disease. Biochem J 2020; 477:833-852. [PMID: 32108870 PMCID: PMC7054746 DOI: 10.1042/bcj20190872] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/14/2020] [Accepted: 01/31/2020] [Indexed: 12/11/2022]
Abstract
Prion diseases are fatal transmissible neurodegenerative conditions of humans and animals that arise through neurotoxicity induced by PrP misfolding. The cellular and molecular mechanisms of prion-induced neurotoxicity remain undefined. Understanding these processes will underpin therapeutic and control strategies for human and animal prion diseases, respectively. Prion diseases are difficult to study in their natural hosts and require the use of tractable animal models. Here we used RNA-Seq-based transcriptome analysis of prion-exposed Drosophila to probe the mechanism of prion-induced neurotoxicity. Adult Drosophila transgenic for pan neuronal expression of ovine PrP targeted to the plasma membrane exhibit a neurotoxic phenotype evidenced by decreased locomotor activity after exposure to ovine prions at the larval stage. Pathway analysis and quantitative PCR of genes differentially expressed in prion-infected Drosophila revealed up-regulation of cell cycle activity and DNA damage response, followed by down-regulation of eIF2 and mTOR signalling. Mitochondrial dysfunction was identified as the principal toxicity pathway in prion-exposed PrP transgenic Drosophila. The transcriptomic changes we observed were specific to PrP targeted to the plasma membrane since these prion-induced gene expression changes were not evident in similarly treated Drosophila transgenic for cytosolic pan neuronal PrP expression, or in non-transgenic control flies. Collectively, our data indicate that aberrant cell cycle activity, repression of protein synthesis and altered mitochondrial function are key events involved in prion-induced neurotoxicity, and correlate with those identified in mammalian hosts undergoing prion disease. These studies highlight the use of PrP transgenic Drosophila as a genetically well-defined tractable host to study mammalian prion biology.
Collapse
|
18
|
Asante EA, Linehan JM, Tomlinson A, Jakubcova T, Hamdan S, Grimshaw A, Smidak M, Jeelani A, Nihat A, Mead S, Brandner S, Wadsworth JDF, Collinge J. Spontaneous generation of prions and transmissible PrP amyloid in a humanised transgenic mouse model of A117V GSS. PLoS Biol 2020; 18:e3000725. [PMID: 32516343 PMCID: PMC7282622 DOI: 10.1371/journal.pbio.3000725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/06/2020] [Indexed: 12/02/2022] Open
Abstract
Inherited prion diseases are caused by autosomal dominant coding mutations in the human prion protein (PrP) gene (PRNP) and account for about 15% of human prion disease cases worldwide. The proposed mechanism is that the mutation predisposes to conformational change in the expressed protein, leading to the generation of disease-related multichain PrP assemblies that propagate by seeded protein misfolding. Despite considerable experimental support for this hypothesis, to-date spontaneous formation of disease-relevant, transmissible PrP assemblies in transgenic models expressing only mutant human PrP has not been demonstrated. Here, we report findings from transgenic mice that express human PrP 117V on a mouse PrP null background (117VV Tg30 mice), which model the PRNP A117V mutation causing inherited prion disease (IPD) including Gerstmann-Sträussler-Scheinker (GSS) disease phenotypes in humans. By studying brain samples from uninoculated groups of mice, we discovered that some mice (≥475 days old) spontaneously generated abnormal PrP assemblies, which after inoculation into further groups of 117VV Tg30 mice, produced a molecular and neuropathological phenotype congruent with that seen after transmission of brain isolates from IPD A117V patients to the same mice. To the best of our knowledge, the 117VV Tg30 mouse line is the first transgenic model expressing only mutant human PrP to show spontaneous generation of transmissible PrP assemblies that directly mirror those generated in an inherited prion disease in humans. Transgenic mice expressing the human prion protein containing a mutation linked to the inherited prion disease Gerstmann-Sträussler-Scheinker disease develop spontaneous neuropathology. This represents the first human prion protein transgenic model to show spontaneous generation of transmissible prion assemblies that directly mirror those generated in humans.
Collapse
Affiliation(s)
- Emmanuel A. Asante
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
- * E-mail: (EAA); (JDFW); (JC)
| | | | - Andrew Tomlinson
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| | - Tatiana Jakubcova
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| | - Shyma Hamdan
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| | - Andrew Grimshaw
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| | - Michelle Smidak
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| | - Asif Jeelani
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| | - Akin Nihat
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| | - Simon Mead
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| | - Sebastian Brandner
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology and Division of Neuropathology, the National Hospital For Neurology and Neurosurgery, University College London NHS Foundation Trust, Queen Square, London United Kingdom
| | - Jonathan D. F. Wadsworth
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
- * E-mail: (EAA); (JDFW); (JC)
| | - John Collinge
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
- * E-mail: (EAA); (JDFW); (JC)
| |
Collapse
|
19
|
Abstract
Mammalian prion diseases are a group of neurodegenerative conditions caused by infection of the central nervous system with proteinaceous agents called prions, including sporadic, variant, and iatrogenic Creutzfeldt-Jakob disease; kuru; inherited prion disease; sheep scrapie; bovine spongiform encephalopathy; and chronic wasting disease. Prions are composed of misfolded and multimeric forms of the normal cellular prion protein (PrP). Prion diseases require host expression of the prion protein gene (PRNP) and a range of other cellular functions to support their propagation and toxicity. Inherited forms of prion disease are caused by mutation of PRNP, whereas acquired and sporadically occurring mammalian prion diseases are controlled by powerful genetic risk and modifying factors. Whereas some PrP amino acid variants cause the disease, others confer protection, dramatically altered incubation times, or changes in the clinical phenotype. Multiple mechanisms, including interference with homotypic protein interactions and the selection of the permissible prion strains in a host, play a role. Several non-PRNP factors have now been uncovered that provide insights into pathways of disease susceptibility or neurotoxicity.
Collapse
Affiliation(s)
- Simon Mead
- Medical Research Council Prion Unit at UCL, Institute of Prion Diseases, University College London, London W1W 7FF, United Kingdom;
| | - Sarah Lloyd
- Medical Research Council Prion Unit at UCL, Institute of Prion Diseases, University College London, London W1W 7FF, United Kingdom;
| | - John Collinge
- Medical Research Council Prion Unit at UCL, Institute of Prion Diseases, University College London, London W1W 7FF, United Kingdom;
| |
Collapse
|
20
|
Chiesa R, Restelli E, Comerio L, Del Gallo F, Imeri L. Transgenic mice recapitulate the phenotypic heterogeneity of genetic prion diseases without developing prion infectivity: Role of intracellular PrP retention in neurotoxicity. Prion 2017; 10:93-102. [PMID: 26864450 PMCID: PMC4981194 DOI: 10.1080/19336896.2016.1139276] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Genetic prion diseases are degenerative brain disorders caused by mutations in the gene encoding the prion protein (PrP). Different PrP mutations cause different diseases, including Creutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker (GSS) syndrome and fatal familial insomnia (FFI). The reason for this variability is not known. It has been suggested that prion strains with unique self-replicating and neurotoxic properties emerge spontaneously in individuals carrying PrP mutations, dictating the phenotypic expression of disease. We generated transgenic mice expressing the FFI mutation, and found that they developed a fatal neurological illness highly reminiscent of FFI, and different from those of similarly generated mice modeling genetic CJD and GSS. Thus transgenic mice recapitulate the phenotypic differences seen in humans. The mutant PrPs expressed in these mice are misfolded but unable to self-replicate. They accumulate in different compartments of the neuronal secretory pathway, impairing the membrane delivery of ion channels essential for neuronal function. Our results indicate that conversion of mutant PrP into an infectious isoform is not required for pathogenesis, and suggest that the phenotypic variability may be due to different effects of mutant PrP on intracellular transport.
Collapse
Affiliation(s)
- Roberto Chiesa
- a Laboratory of Prion Neurobiology, Department of Neuroscience, IRCCS - "Mario Negri" Institute for Pharmacological Research , Milan , Italy
| | - Elena Restelli
- a Laboratory of Prion Neurobiology, Department of Neuroscience, IRCCS - "Mario Negri" Institute for Pharmacological Research , Milan , Italy
| | - Liliana Comerio
- a Laboratory of Prion Neurobiology, Department of Neuroscience, IRCCS - "Mario Negri" Institute for Pharmacological Research , Milan , Italy
| | - Federico Del Gallo
- b Department of Health Sciences , University of Milan Medical School , Milan , Italy
| | - Luca Imeri
- b Department of Health Sciences , University of Milan Medical School , Milan , Italy
| |
Collapse
|
21
|
Watts JC, Prusiner SB. Experimental Models of Inherited PrP Prion Diseases. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a027151. [PMID: 28096244 DOI: 10.1101/cshperspect.a027151] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The inherited prion protein (PrP) prion disorders, which include familial Creutzfeldt-Jakob disease, Gerstmann-Sträussler-Scheinker disease, and fatal familial insomnia, constitute ∼10%-15% of all PrP prion disease cases in humans. Attempts to generate animal models of these disorders using transgenic mice expressing mutant PrP have produced variable results. Although many lines of mice develop spontaneous signs of neurological illness with accompanying prion disease-specific neuropathological changes, others do not. Furthermore, demonstrating the presence of protease-resistant PrP species and prion infectivity-two of the hallmarks of the PrP prion disorders-in the brains of spontaneously sick mice has proven particularly challenging. Here, we review the progress that has been made toward developing accurate mouse models of the inherited PrP prion disorders.
Collapse
Affiliation(s)
- Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Biochemistry, University of Toronto, Toronto, Ontario M5T 2S8, Canada
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, Departments of Neurology and Biochemistry and Biophysics, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
22
|
Genetic human prion disease modelled in PrP transgenic Drosophila. Biochem J 2017; 474:3253-3267. [PMID: 28814578 PMCID: PMC5606059 DOI: 10.1042/bcj20170462] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/07/2017] [Accepted: 08/15/2017] [Indexed: 12/26/2022]
Abstract
Inherited human prion diseases, such as fatal familial insomnia (FFI) and familial Creutzfeldt–Jakob disease (fCJD), are associated with autosomal dominant mutations in the human prion protein gene PRNP and accumulation of PrPSc, an abnormal isomer of the normal host protein PrPC, in the brain of affected individuals. PrPSc is the principal component of the transmissible neurotoxic prion agent. It is important to identify molecular pathways and cellular processes that regulate prion formation and prion-induced neurotoxicity. This will allow identification of possible therapeutic interventions for individuals with, or at risk from, genetic human prion disease. Increasingly, Drosophila has been used to model human neurodegenerative disease. An important unanswered question is whether genetic prion disease with concomitant spontaneous prion formation can be modelled in Drosophila. We have used pUAST/PhiC31-mediated site-directed mutagenesis to generate Drosophila transgenic for murine or hamster PrP (prion protein) that carry single-codon mutations associated with genetic human prion disease. Mouse or hamster PrP harbouring an FFI (D178N) or fCJD (E200K) mutation showed mild Proteinase K resistance when expressed in Drosophila. Adult Drosophila transgenic for FFI or fCJD variants of mouse or hamster PrP displayed a spontaneous decline in locomotor ability that increased in severity as the flies aged. Significantly, this mutant PrP-mediated neurotoxic fly phenotype was transferable to recipient Drosophila that expressed the wild-type form of the transgene. Collectively, our novel data are indicative of the spontaneous formation of a PrP-dependent neurotoxic phenotype in FFI- or CJD-PrP transgenic Drosophila and show that inherited human prion disease can be modelled in this invertebrate host.
Collapse
|
23
|
Elezgarai SR, Fernández-Borges N, Eraña H, Sevillano AM, Charco JM, Harrathi C, Saá P, Gil D, Kong Q, Requena JR, Andréoletti O, Castilla J. Generation of a new infectious recombinant prion: a model to understand Gerstmann-Sträussler-Scheinker syndrome. Sci Rep 2017; 7:9584. [PMID: 28851967 PMCID: PMC5575253 DOI: 10.1038/s41598-017-09489-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/10/2017] [Indexed: 12/15/2022] Open
Abstract
Human transmissible spongiform encephalopathies (TSEs) or prion diseases are a group of fatal neurodegenerative disorders that include Kuru, Creutzfeldt-Jakob disease, Gerstmann-Sträussler-Scheinker syndrome (GSS), and fatal familial insomnia. GSS is a genetically determined TSE caused by a range of mutations within the prion protein (PrP) gene. Several animal models, based on the expression of PrPs carrying mutations analogous to human heritable prion diseases, support that mutations might predispose PrP to spontaneously misfold. An adapted Protein Misfolding Cyclic Amplification methodology based on the use of human recombinant PrP (recPMCA) generated different self-propagating misfolded proteins spontaneously. These were characterized biochemically and structurally, and the one partially sharing some of the GSS PrPSc molecular features was inoculated into different animal models showing high infectivity. This constitutes an infectious recombinant prion which could be an invaluable model for understanding GSS. Moreover, this study proves the possibility to generate recombinant versions of other human prion diseases that could provide a further understanding on the molecular features of these devastating disorders.
Collapse
Affiliation(s)
- Saioa R Elezgarai
- CIC bioGUNE, Parque tecnológico de Bizkaia, Derio, 48160, Bizkaia, Spain
| | | | - Hasier Eraña
- CIC bioGUNE, Parque tecnológico de Bizkaia, Derio, 48160, Bizkaia, Spain
| | - Alejandro M Sevillano
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Jorge M Charco
- CIC bioGUNE, Parque tecnológico de Bizkaia, Derio, 48160, Bizkaia, Spain
| | - Chafik Harrathi
- CIC bioGUNE, Parque tecnológico de Bizkaia, Derio, 48160, Bizkaia, Spain
| | - Paula Saá
- American Red Cross, Gaithersburg, MD, USA
| | - David Gil
- CIC bioGUNE, Parque tecnológico de Bizkaia, Derio, 48160, Bizkaia, Spain
| | - Qingzhong Kong
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jesús R Requena
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Olivier Andréoletti
- Ecole Nationale du Veterinaire, Service de Pathologie du Bétail, Toulouse, 31076, France
| | - Joaquín Castilla
- CIC bioGUNE, Parque tecnológico de Bizkaia, Derio, 48160, Bizkaia, Spain. .,IKERBASQUE, Basque Foundation for Science, Bilbao, 48011, Bizkaia, Spain.
| |
Collapse
|
24
|
Diack AB, Alibhai JD, Manson JC. Gene Targeted Transgenic Mouse Models in Prion Research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:157-179. [PMID: 28838660 DOI: 10.1016/bs.pmbts.2017.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The production of transgenic mice expressing different forms of the prion protein (PrP) or devoid of PrP has enabled researchers to study the role of PrP in the infectious process of a prion disease and its normal function in the healthy individual. A wide range of transgenic models have been produced ranging from PrP null mice, normal expression levels to overexpression models, models expressing different species of the Prnp gene and different mutations and polymorphisms within the gene. Using this range of transgenic models has allowed us to define the influence of PrP expression on disease susceptibility and transmission, assess zoonotic potential, define strains of human prion diseases, elucidate the function of PrP, and start to unravel the mechanisms involved in chronic neurodegeneration. This chapter focuses mainly on the use of the gene targeted transgenic models and summarizes the ways in which they have allowed us to study the role of PrP in prion disease and the insights they have provided into the mechanisms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Abigail B Diack
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, United Kingdom.
| | - James D Alibhai
- The National CJD Research and Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jean C Manson
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, United Kingdom
| |
Collapse
|
25
|
Eaton SL, Wishart TM. Bridging the gap: large animal models in neurodegenerative research. Mamm Genome 2017; 28:324-337. [PMID: 28378063 PMCID: PMC5569151 DOI: 10.1007/s00335-017-9687-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/25/2017] [Indexed: 01/08/2023]
Abstract
The world health organisation has declared neurological disorders as one of the greatest public health risks in the world today. Yet, despite this growing concern, the mechanisms underpinning many of these conditions are still poorly understood. This may in part be due to the seemingly diverse nature of the initiating insults ranging from genetic (such as the Ataxia's and Lysosomal storage disorders) through to protein misfolding and aggregation (i.e. Prions), and those of a predominantly unknown aetiology (i.e. Alzheimer's and Parkinson's disease). However, efforts to elucidate mechanistic regulation are also likely to be hampered because of the complexity of the human nervous system, the apparent selective regional vulnerability and differential degenerative progression. The key to elucidating these aetiologies is determining the regional molecular cascades, which are occurring from the early through to terminal stages of disease progression. Whilst much molecular data have been captured at the end stage of disease from post-mortem analysis in humans, the very early stages of disease are often conspicuously asymptomatic, and even if they were not, repeated sampling from multiple brain regions of "affected" patients and "controls" is neither ethical nor possible. Model systems therefore become fundamental for elucidating the mechanisms governing these complex neurodegenerative conditions. However, finding a model that precisely mimics the human condition can be challenging and expensive. Whilst cellular and invertebrate models are frequently used in neurodegenerative research and have undoubtedly yielded much useful data, the comparatively simplistic nature of these systems makes insights gained from such a stand alone model limited when it comes to translation. Given the recent advances in gene editing technology, the options for novel model generation in higher order species have opened up new and exciting possibilities for the field. In this review, we therefore explain some of the reasons why larger animal models often appear to give a more robust recapitulation of human neurological disorders and why they may be a critical stepping stone for effective therapeutic translation.
Collapse
Affiliation(s)
- S L Eaton
- Roslin Institute and Royal (Dick) Veterinary studies, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - T M Wishart
- Roslin Institute and Royal (Dick) Veterinary studies, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK.
- Euan MacDonald Centre for MND Research, Chancellor's Building, 49 Little France, Edinburgh, EH16 4SB, UK.
| |
Collapse
|
26
|
Brandner S, Jaunmuktane Z. Prion disease: experimental models and reality. Acta Neuropathol 2017; 133:197-222. [PMID: 28084518 PMCID: PMC5250673 DOI: 10.1007/s00401-017-1670-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 01/04/2023]
Abstract
The understanding of the pathogenesis and mechanisms of diseases requires a multidisciplinary approach, involving clinical observation, correlation to pathological processes, and modelling of disease mechanisms. It is an inherent challenge, and arguably impossible to generate model systems that can faithfully recapitulate all aspects of human disease. It is, therefore, important to be aware of the potentials and also the limitations of specific model systems. Model systems are usually designed to recapitulate only specific aspects of the disease, such as a pathological phenotype, a pathomechanism, or to test a hypothesis. Here, we evaluate and discuss model systems that were generated to understand clinical, pathological, genetic, biochemical, and epidemiological aspects of prion diseases. Whilst clinical research and studies on human tissue are an essential component of prion research, much of the understanding of the mechanisms governing transmission, replication, and toxicity comes from in vitro and in vivo studies. As with other neurodegenerative diseases caused by protein misfolding, the pathogenesis of prion disease is complex, full of conundra and contradictions. We will give here a historical overview of the use of models of prion disease, how they have evolved alongside the scientific questions, and how advancements in technologies have pushed the boundaries of our understanding of prion biology.
Collapse
Affiliation(s)
- Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London, WC1N 3BG UK
| | - Zane Jaunmuktane
- Department of Neurodegenerative Disease, UCL Institute of Neurology and Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London, WC1N 3BG UK
| |
Collapse
|
27
|
Moreno JA, Telling GC. Insights into Mechanisms of Transmission and Pathogenesis from Transgenic Mouse Models of Prion Diseases. Methods Mol Biol 2017; 1658:219-252. [PMID: 28861793 DOI: 10.1007/978-1-4939-7244-9_16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Prions represent a new paradigm of protein-mediated information transfer. In the case of mammals, prions are the cause of fatal, transmissible neurodegenerative diseases, sometimes referred to as transmissible spongiform encephalopathies (TSEs), which frequently occur as epidemics. An increasing body of evidence indicates that the canonical mechanism of conformational corruption of cellular prion protein (PrPC) by the pathogenic isoform (PrPSc) that is the basis of prion formation in TSEs is common to a spectrum of proteins associated with various additional human neurodegenerative disorders, including the more common Alzheimer's and Parkinson's diseases. The peerless infectious properties of TSE prions, and the unparalleled tools for their study, therefore enable elucidation of mechanisms of template-mediated conformational propagation that are generally applicable to these related disease states. Many unresolved issues remain including the exact molecular nature of the prion, the detailed cellular and molecular mechanisms of prion propagation, and the means by which prion diseases can be both genetic and infectious. In addition, we know little about the mechanism by which neurons degenerate during prion diseases. Tied to this, the physiological role of the normal form of the prion protein remains unclear and it is uncertain whether or not loss of this function contributes to prion pathogenesis. The factors governing the transmission of prions between species remain unclear, in particular the means by which prion strains and PrP primary structure interact to affect interspecies prion transmission. Despite all these unknowns, advances in our understanding of prions have occurred because of their transmissibility to experimental animals, and the development of transgenic (Tg) mouse models has done much to further our understanding about various aspects of prion biology. In this review, we will focus on advances in our understanding of prion biology that occurred in the past 8 years since our last review of this topic.
Collapse
Affiliation(s)
- Julie A Moreno
- Cell and Molecular Biology Graduate Program, Molecular, Cellular and Integrative Neuroscience Graduate Program, Department of Microbiology, Immunology and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, 80523, USA
| | - Glenn C Telling
- Cell and Molecular Biology Graduate Program, Molecular, Cellular and Integrative Neuroscience Graduate Program, Department of Microbiology, Immunology and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, 80523, USA.
| |
Collapse
|
28
|
Mehrabian M, Hildebrandt H, Schmitt-Ulms G. NCAM1 Polysialylation: The Prion Protein's Elusive Reason for Being? ASN Neuro 2016; 8:8/6/1759091416679074. [PMID: 27879349 PMCID: PMC5122176 DOI: 10.1177/1759091416679074] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 09/08/2016] [Accepted: 10/02/2016] [Indexed: 01/06/2023] Open
Abstract
Much confusion surrounds the physiological function of the cellular prion protein (PrPC). It is, however, anticipated that knowledge of its function will shed light on its contribution to neurodegenerative diseases and suggest ways to interfere with the cellular toxicity central to them. Consequently, efforts to elucidate its function have been all but exhaustive. Building on earlier work that uncovered the evolutionary descent of the prion founder gene from an ancestral ZIP zinc transporter, we recently investigated a possible role of PrPC in a morphogenetic program referred to as epithelial-to-mesenchymal transition (EMT). By capitalizing on PrPC knockout cell clones in a mammalian cell model of EMT and using a comparative proteomics discovery strategy, neural cell adhesion molecule-1 emerged as a protein whose upregulation during EMT was perturbed in PrPC knockout cells. Follow-up work led us to observe that PrPC regulates the polysialylation of the neural cell adhesion molecule NCAM1 in cells undergoing morphogenetic reprogramming. In addition to governing cellular migration, polysialylation modulates several other cellular plasticity programs PrPC has been phenotypically linked to. These include neurogenesis in the subventricular zone, controlled mossy fiber sprouting and trimming in the hippocampal formation, hematopoietic stem cell renewal, myelin repair and maintenance, integrity of the circadian rhythm, and glutamatergic signaling. This review revisits this body of literature and attempts to present it in light of this novel contextual framework. When approached in this manner, a coherent model of PrPC acting as a regulator of polysialylation during specific cell and tissue morphogenesis events comes into focus.
Collapse
Affiliation(s)
- Mohadeseh Mehrabian
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Herbert Hildebrandt
- Institute for Cellular Chemistry, Hannover Medical School, Hannover, Germany
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Minikel EV, Vallabh SM, Lek M, Estrada K, Samocha KE, Sathirapongsasuti JF, McLean CY, Tung JY, Yu LPC, Gambetti P, Blevins J, Zhang S, Cohen Y, Chen W, Yamada M, Hamaguchi T, Sanjo N, Mizusawa H, Nakamura Y, Kitamoto T, Collins SJ, Boyd A, Will RG, Knight R, Ponto C, Zerr I, Kraus TFJ, Eigenbrod S, Giese A, Calero M, de Pedro-Cuesta J, Haïk S, Laplanche JL, Bouaziz-Amar E, Brandel JP, Capellari S, Parchi P, Poleggi A, Ladogana A, O'Donnell-Luria AH, Karczewski KJ, Marshall JL, Boehnke M, Laakso M, Mohlke KL, Kähler A, Chambert K, McCarroll S, Sullivan PF, Hultman CM, Purcell SM, Sklar P, van der Lee SJ, Rozemuller A, Jansen C, Hofman A, Kraaij R, van Rooij JGJ, Ikram MA, Uitterlinden AG, van Duijn CM, Daly MJ, MacArthur DG. Quantifying prion disease penetrance using large population control cohorts. Sci Transl Med 2016; 8:322ra9. [PMID: 26791950 DOI: 10.1126/scitranslmed.aad5169] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
More than 100,000 genetic variants are reported to cause Mendelian disease in humans, but the penetrance-the probability that a carrier of the purported disease-causing genotype will indeed develop the disease-is generally unknown. We assess the impact of variants in the prion protein gene (PRNP) on the risk of prion disease by analyzing 16,025 prion disease cases, 60,706 population control exomes, and 531,575 individuals genotyped by 23andMe Inc. We show that missense variants in PRNP previously reported to be pathogenic are at least 30 times more common in the population than expected on the basis of genetic prion disease prevalence. Although some of this excess can be attributed to benign variants falsely assigned as pathogenic, other variants have genuine effects on disease susceptibility but confer lifetime risks ranging from <0.1 to ~100%. We also show that truncating variants in PRNP have position-dependent effects, with true loss-of-function alleles found in healthy older individuals, a finding that supports the safety of therapeutic suppression of prion protein expression.
Collapse
Affiliation(s)
- Eric Vallabh Minikel
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA. Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA. Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA. Prion Alliance, Cambridge, MA 02139, USA.
| | - Sonia M Vallabh
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA. Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA. Prion Alliance, Cambridge, MA 02139, USA
| | - Monkol Lek
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA. Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Karol Estrada
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA. Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kaitlin E Samocha
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA. Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA. Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | | | - Cory Y McLean
- Research, 23andMe Inc., Mountain View, CA 94041, USA
| | - Joyce Y Tung
- Research, 23andMe Inc., Mountain View, CA 94041, USA
| | - Linda P C Yu
- Research, 23andMe Inc., Mountain View, CA 94041, USA
| | - Pierluigi Gambetti
- National Prion Disease Pathology Surveillance Center, Cleveland, OH 44106, USA
| | - Janis Blevins
- National Prion Disease Pathology Surveillance Center, Cleveland, OH 44106, USA
| | - Shulin Zhang
- University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Yvonne Cohen
- National Prion Disease Pathology Surveillance Center, Cleveland, OH 44106, USA
| | - Wei Chen
- National Prion Disease Pathology Surveillance Center, Cleveland, OH 44106, USA
| | - Masahito Yamada
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - Tsuyoshi Hamaguchi
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - Nobuo Sanjo
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Hidehiro Mizusawa
- National Center Hospital, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan
| | - Yosikazu Nakamura
- Department of Public Health, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Tetsuyuki Kitamoto
- Department of Neurological Science, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Steven J Collins
- Australian National Creutzfeldt-Jakob Disease Registry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alison Boyd
- Australian National Creutzfeldt-Jakob Disease Registry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Robert G Will
- National Creutzfeldt-Jakob Disease Research & Surveillance Unit, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Richard Knight
- National Creutzfeldt-Jakob Disease Research & Surveillance Unit, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Claudia Ponto
- National Reference Center for the Surveillance of Human Transmissible Spongiform Encephalopathies, Georg-August-University, Goettingen 37073, Germany
| | - Inga Zerr
- National Reference Center for the Surveillance of Human Transmissible Spongiform Encephalopathies, Georg-August-University, Goettingen 37073, Germany
| | - Theo F J Kraus
- Center for Neuropathology and Prion Research (ZNP), Ludwig-Maximilians-University, Munich 81377, Germany
| | - Sabina Eigenbrod
- Center for Neuropathology and Prion Research (ZNP), Ludwig-Maximilians-University, Munich 81377, Germany
| | - Armin Giese
- Center for Neuropathology and Prion Research (ZNP), Ludwig-Maximilians-University, Munich 81377, Germany
| | - Miguel Calero
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid 28031, Spain
| | - Jesús de Pedro-Cuesta
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid 28031, Spain
| | - Stéphane Haïk
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, Pierre and Marie Curie University Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Epinière, 75013 Paris, France. Assistance Publique-Hôpitaux de Paris (AP-HP), Cellule Nationale de Référence des Maladies de Creutzfeldt-Jakob, Groupe Hospitalier Pitié-Salpêtrière, F-75013 Paris, France
| | - Jean-Louis Laplanche
- AP-HP, Service de Biochimie et Biologie Moléculaire, Hôpital Lariboisière, 75010 Paris, France
| | - Elodie Bouaziz-Amar
- AP-HP, Service de Biochimie et Biologie Moléculaire, Hôpital Lariboisière, 75010 Paris, France
| | - Jean-Philippe Brandel
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, Pierre and Marie Curie University Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Epinière, 75013 Paris, France. Assistance Publique-Hôpitaux de Paris (AP-HP), Cellule Nationale de Référence des Maladies de Creutzfeldt-Jakob, Groupe Hospitalier Pitié-Salpêtrière, F-75013 Paris, France
| | - Sabina Capellari
- Istituto di Ricovero e Cura a Carattere Scientifico, Institute of Neurological Sciences, Bologna 40123, Italy. Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40126, Italy
| | - Piero Parchi
- Istituto di Ricovero e Cura a Carattere Scientifico, Institute of Neurological Sciences, Bologna 40123, Italy. Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40126, Italy
| | - Anna Poleggi
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Anna Ladogana
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Anne H O'Donnell-Luria
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA. Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA. Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Konrad J Karczewski
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA. Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jamie L Marshall
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA. Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio 70210, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Anna Kähler
- Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Kimberly Chambert
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Steven McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Patrick F Sullivan
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA. Karolinska Institutet, Stockholm SE-171 77, Sweden
| | | | - Shaun M Purcell
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pamela Sklar
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sven J van der Lee
- Department of Epidemiology, Erasmus Medical Center (MC), Rotterdam 3000 CA, Netherlands
| | - Annemieke Rozemuller
- Dutch Surveillance Centre for Prion Diseases, Department of Pathology, University Medical Center, Utrecht 3584 CX, Netherlands
| | - Casper Jansen
- Dutch Surveillance Centre for Prion Diseases, Department of Pathology, University Medical Center, Utrecht 3584 CX, Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center (MC), Rotterdam 3000 CA, Netherlands
| | - Robert Kraaij
- Department of Internal Medicine, Erasmus MC, Rotterdam 3000 CA, Netherlands
| | | | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center (MC), Rotterdam 3000 CA, Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center (MC), Rotterdam 3000 CA, Netherlands. Department of Internal Medicine, Erasmus MC, Rotterdam 3000 CA, Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus Medical Center (MC), Rotterdam 3000 CA, Netherlands
| | | | - Mark J Daly
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA. Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Daniel G MacArthur
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA. Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
30
|
Watts JC, Giles K, Bourkas MEC, Patel S, Oehler A, Gavidia M, Bhardwaj S, Lee J, Prusiner SB. Towards authentic transgenic mouse models of heritable PrP prion diseases. Acta Neuropathol 2016; 132:593-610. [PMID: 27350609 DOI: 10.1007/s00401-016-1585-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/24/2016] [Accepted: 05/27/2016] [Indexed: 11/27/2022]
Abstract
Attempts to model inherited human prion disorders such as familial Creutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker (GSS) disease, and fatal familial insomnia (FFI) using genetically modified mice have produced disappointing results. We recently demonstrated that transgenic (Tg) mice expressing wild-type bank vole prion protein (BVPrP) containing isoleucine at polymorphic codon 109 develop a spontaneous neurodegenerative disorder that exhibits many of the hallmarks of prion disease. To determine if mutations causing inherited human prion disease alter this phenotype, we generated Tg mice expressing BVPrP containing the D178N mutation, which causes FFI; the E200K mutation, which causes familial CJD; or an anchorless PrP mutation similar to mutations that cause GSS. Modest expression levels of mutant BVPrP resulted in highly penetrant spontaneous disease in Tg mice, with mean ages of disease onset ranging from ~120 to ~560 days. The brains of spontaneously ill mice exhibited prominent features of prion disease-specific neuropathology that were unique to each mutation and distinct from Tg mice expressing wild-type BVPrP. An ~8-kDa proteinase K-resistant PrP fragment was found in the brains of spontaneously ill Tg mice expressing either wild-type or mutant BVPrP. The spontaneously formed mutant BVPrP prions were transmissible to Tg mice expressing wild-type or mutant BVPrP as well as to Tg mice expressing mouse PrP. Thus, Tg mice expressing mutant BVPrP exhibit many of the hallmarks of heritable prion disorders in humans including spontaneous disease, protease-resistant PrP, and prion infectivity.
Collapse
Affiliation(s)
- Joel C Watts
- Institute for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94143-0518, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Biochemistry, University of Toronto, Toronto, ON, M5T 2S8, Canada
| | - Kurt Giles
- Institute for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94143-0518, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Matthew E C Bourkas
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Biochemistry, University of Toronto, Toronto, ON, M5T 2S8, Canada
| | - Smita Patel
- Institute for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94143-0518, USA
| | - Abby Oehler
- Institute for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94143-0518, USA
| | - Marta Gavidia
- Institute for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94143-0518, USA
| | - Sumita Bhardwaj
- Institute for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94143-0518, USA
| | - Joanne Lee
- Institute for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94143-0518, USA
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94143-0518, USA.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
31
|
Kaczmarczyk L, Mende Y, Zevnik B, Jackson WS. Manipulating the Prion Protein Gene Sequence and Expression Levels with CRISPR/Cas9. PLoS One 2016; 11:e0154604. [PMID: 27128441 PMCID: PMC4851410 DOI: 10.1371/journal.pone.0154604] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 04/16/2016] [Indexed: 01/01/2023] Open
Abstract
The mammalian prion protein (PrP, encoded by Prnp) is most infamous for its central role in prion diseases, invariably fatal neurodegenerative diseases affecting humans, food animals, and animals in the wild. However, PrP is also hypothesized to be an important receptor for toxic protein conformers in Alzheimer's disease, and is associated with other clinically relevant processes such as cancer and stroke. Thus, key insights into important clinical areas, as well as into understanding PrP functions in normal physiology, can be obtained from studying transgenic mouse models and cell culture systems. However, the Prnp locus is difficult to manipulate by homologous recombination, making modifications of the endogenous locus rarely attempted. Fortunately in recent years genome engineering technologies, like TALENs or CRISPR/Cas9 (CC9), have brought exceptional new possibilities for manipulating Prnp. Herein, we present our observations made during systematic experiments with the CC9 system targeting the endogenous mouse Prnp locus, to either modify sequences or to boost PrP expression using CC9-based synergistic activation mediators (SAMs). It is our hope that this information will aid and encourage researchers to implement gene-targeting techniques into their research program.
Collapse
Affiliation(s)
- Lech Kaczmarczyk
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ylva Mende
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), Medical Faculty, University of Cologne, Cologne, Germany
| | - Branko Zevnik
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), Medical Faculty, University of Cologne, Cologne, Germany
| | - Walker S. Jackson
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- * E-mail:
| |
Collapse
|
32
|
Brandel JP, Haïk S. Malattie da prioni o encefalopatie spongiformi trasmissibili. Neurologia 2016. [DOI: 10.1016/s1634-7072(16)77562-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
33
|
Brandel JP, Corbillé AG, Derkinderen P, Haïk S. [Is Parkinson's disease a prion disease?]. Rev Neurol (Paris) 2015; 171:812-24. [PMID: 26563663 PMCID: PMC7111738 DOI: 10.1016/j.neurol.2015.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 02/07/2023]
Abstract
The accumulation of a specific protein in aggregated form is a common phenomenon in human neurodegenerative diseases. In Parkinson's disease, this protein is α-synuclein which is a neuronal protein of 143 amino acids. With a monomeric conformation in solution, it also has a natural capacity to aggregate into amyloid structures (dimers, oligomers, fibrils and Lewy bodies or neurites). It therefore fulfils the characteristics of a prion protein (different conformations, seeding and spreading). In vitro and in vivo experimental evidence in transgenic and wild animals indicates a prion-like propagation of Parkinson's disease. The sequential and predictive distribution of α-synuclein demonstrated by Braak et al. and its correlation with non-motor signs are consistent with the prion-like progression. Although the triggering factor causing the misfolding and aggregation of the target protein is unknown, Parkinson's disease is a highly relevant model for the study of these mechanisms and also to test specific treatments targeting the assemblies of α-synuclein and propagation from pre-motor phase of the disease. Despite this prion-like progression, there is currently no argument indicating a risk of human transmission of Parkinson's disease.
Collapse
Affiliation(s)
- J-P Brandel
- Inserm U 1127, CNRS UMR 7225, Sorbonne universités, UPMC University Paris 06 UMR S 1127, institut du cerveau et de la mœlle épinière, ICM, 75013 Paris, France; Cellule nationale de référence des maladies de Creutzfeldt-Jakob, groupe hospitalier Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75651 Paris cedex 13, France; Centre national de référence des agents transmissibles non conventionnels, 75013 Paris, France; Unité James-Parkinson, Fondation ophtalmologique Rothschild, 75019 Paris, France.
| | - A-G Corbillé
- Département de neurologie, CHU de Nantes, 44093 Nantes, France; Inserm, U913, 44093 Nantes, France
| | - P Derkinderen
- Département de neurologie, CHU de Nantes, 44093 Nantes, France; Inserm, U913, 44093 Nantes, France
| | - S Haïk
- Inserm U 1127, CNRS UMR 7225, Sorbonne universités, UPMC University Paris 06 UMR S 1127, institut du cerveau et de la mœlle épinière, ICM, 75013 Paris, France; Cellule nationale de référence des maladies de Creutzfeldt-Jakob, groupe hospitalier Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75651 Paris cedex 13, France; Centre national de référence des agents transmissibles non conventionnels, 75013 Paris, France
| |
Collapse
|
34
|
Watts JC, Giles K, Serban A, Patel S, Oehler A, Bhardwaj S, Guan S, Greicius MD, Miller BL, DeArmond SJ, Geschwind MD, Prusiner SB. Modulation of Creutzfeldt-Jakob disease prion propagation by the A224V mutation. Ann Neurol 2015; 78:540-53. [PMID: 26094969 DOI: 10.1002/ana.24463] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 06/17/2015] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Mutations in the gene encoding the prion protein (PrP) are responsible for approximately 10 to 15% of cases of prion disease in humans, including Creutzfeldt-Jakob disease (CJD). Here, we report on the discovery of a previously unreported C-terminal PrP mutation (A224V) in a CJD patient exhibiting a disease similar to the rare VV1 subtype of sporadic (s) CJD and investigate the role of this mutation in prion replication and transmission. METHODS We generated transgenic (Tg) mice expressing human PrP with the V129 polymorphism and A224V mutation, denoted Tg(HuPrP,V129,A224V) mice, and inoculated them with different subtypes of sCJD prions. RESULTS Transmission of sCJD VV2 or MV2 prions was accelerated in Tg(HuPrP,V129,A224V) mice, compared to Tg(HuPrP,V129) mice, with incubation periods of ∼110 and ∼210 days, respectively. In contrast, sCJD MM1 prions resulted in longer incubation periods in Tg(HuPrP,V129,A224V) mice, compared to Tg(HuPrP,V129) mice (∼320 vs. ∼210 days). Prion strain fidelity was maintained in Tg(HuPrP,V129,A224V) mice inoculated with sCJD VV2 or MM1 prions, despite the altered replication kinetics. INTERPRETATION Our results suggest that A224V is a risk factor for prion disease and modulates the transmission behavior of CJD prions in a strain-specific manner, arguing that residues near the C-terminus of PrP are important for controlling the kinetics of prion replication.
Collapse
Affiliation(s)
- Joel C Watts
- Institute for Neurodegenerative Diseases.,Departments of Neurology
| | - Kurt Giles
- Institute for Neurodegenerative Diseases.,Departments of Neurology
| | - Ana Serban
- Institute for Neurodegenerative Diseases
| | | | | | | | - Shenheng Guan
- Institute for Neurodegenerative Diseases.,Pharmaceutical Chemistry
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA
| | | | | | | | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases.,Departments of Neurology.,Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
| |
Collapse
|
35
|
Bujdoso R, Landgraf M, Jackson WS, Thackray AM. Prion-induced neurotoxicity: Possible role for cell cycle activity and DNA damage response. World J Virol 2015; 4:188-197. [PMID: 26279981 PMCID: PMC4534811 DOI: 10.5501/wjv.v4.i3.188] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 03/19/2015] [Accepted: 04/30/2015] [Indexed: 02/05/2023] Open
Abstract
Protein misfolding neurodegenerative diseases arise through neurotoxicity induced by aggregation of host proteins. These conditions include Alzheimer’s disease, Huntington’s disease, Parkinson’s disease, motor neuron disease, tauopathies and prion diseases. Collectively, these conditions are a challenge to society because of the increasing aged population and through the real threat to human food security by animal prion diseases. It is therefore important to understand the cellular and molecular mechanisms that underlie protein misfolding-induced neurotoxicity as this will form the basis for designing strategies to alleviate their burden. Prion diseases are an important paradigm for neurodegenerative conditions in general since several of these maladies have now been shown to display prion-like phenomena. Increasingly, cell cycle activity and the DNA damage response are recognised as cellular events that participate in the neurotoxic process of various neurodegenerative diseases, and their associated animal models, which suggests they are truly involved in the pathogenic process and are not merely epiphenomena. Here we review the role of cell cycle activity and the DNA damage response in neurodegeneration associated with protein misfolding diseases, and suggest that these events contribute towards prion-induced neurotoxicity. In doing so, we highlight PrP transgenic Drosophila as a tractable model for the genetic analysis of transmissible mammalian prion disease.
Collapse
|
36
|
Noble GP, Walsh DJ, Miller MB, Jackson WS, Supattapone S. Requirements for mutant and wild-type prion protein misfolding in vitro. Biochemistry 2015; 54:1180-7. [PMID: 25584902 DOI: 10.1021/bi501495j] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Misfolding of the prion protein (PrP) plays a central role in the pathogenesis of infectious, sporadic, and inherited prion diseases. Here we use a chemically defined prion propagation system to study misfolding of the pathogenic PrP mutant D177N in vitro. This mutation causes PrP to misfold spontaneously in the absence of cofactor molecules in a process dependent on time, temperature, pH, and intermittent sonication. Spontaneously misfolded mutant PrP is able to template its unique conformation onto wild-type PrP substrate in a process that requires a phospholipid activity distinct from that required for the propagation of infectious prions. Similar results were obtained with a second pathogenic PrP mutant, E199K, but not with the polymorphic substitution M128V. Moreover, wild-type PrP inhibits mutant PrP misfolding in a dose-dependent manner, and cofactor molecules can antagonize this effect. These studies suggest that interactions between mutant PrP, wild-type PrP, and other cellular factors may control the rate of PrP misfolding in inherited prion diseases.
Collapse
Affiliation(s)
- Geoffrey P Noble
- Department of Biochemistry, The Geisel School of Medicine at Dartmouth , Vail Building Room 311, Hanover, New Hampshire 03755, United States
| | | | | | | | | |
Collapse
|
37
|
|
38
|
Jackson WS. Selective vulnerability to neurodegenerative disease: the curious case of Prion Protein. Dis Model Mech 2014; 7:21-9. [PMID: 24396151 PMCID: PMC3882045 DOI: 10.1242/dmm.012146] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The mechanisms underlying the selective targeting of specific brain regions by different neurodegenerative diseases is one of the most intriguing mysteries in medicine. For example, it is known that Alzheimer’s disease primarily affects parts of the brain that play a role in memory, whereas Parkinson’s disease predominantly affects parts of the brain that are involved in body movement. However, the reasons that other brain regions remain unaffected in these diseases are unknown. A better understanding of the phenomenon of selective vulnerability is required for the development of targeted therapeutic approaches that specifically protect affected neurons, thereby altering the disease course and preventing its progression. Prion diseases are a fascinating group of neurodegenerative diseases because they exhibit a wide phenotypic spectrum caused by different sequence perturbations in a single protein. The possible ways that mutations affecting this protein can cause several distinct neurodegenerative diseases are explored in this Review to highlight the complexity underlying selective vulnerability. The premise of this article is that selective vulnerability is determined by the interaction of specific protein conformers and region-specific microenvironments harboring unique combinations of subcellular components such as metals, chaperones and protein translation machinery. Given the abundance of potential contributory factors in the neurodegenerative process, a better understanding of how these factors interact will provide invaluable insight into disease mechanisms to guide therapeutic discovery.
Collapse
Affiliation(s)
- Walker S Jackson
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 25, 53127-Bonn, Germany
| |
Collapse
|
39
|
Watts JC, Prusiner SB. Mouse models for studying the formation and propagation of prions. J Biol Chem 2014; 289:19841-9. [PMID: 24860095 DOI: 10.1074/jbc.r114.550707] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Prions are self-propagating protein conformers that cause a variety of neurodegenerative disorders in humans and animals. Mouse models have played key roles in deciphering the biology of prions and in assessing candidate therapeutics. The development of transgenic mice that form prions spontaneously in the brain has advanced our understanding of sporadic and genetic prion diseases. Furthermore, the realization that many proteins can become prions has necessitated the development of mouse models for assessing the potential transmissibility of common neurodegenerative diseases. As the universe of prion diseases continues to expand, mouse models will remain crucial for interrogating these devastating illnesses.
Collapse
Affiliation(s)
- Joel C Watts
- From the Institute for Neurodegenerative Diseases and the Department of Neurology, University of California, San Francisco, California 94143
| | - Stanley B Prusiner
- From the Institute for Neurodegenerative Diseases and the Department of Neurology, University of California, San Francisco, California 94143
| |
Collapse
|
40
|
Translation of the prion protein mRNA is robust in astrocytes but does not amplify during reactive astrocytosis in the mouse brain. PLoS One 2014; 9:e95958. [PMID: 24752288 PMCID: PMC3994155 DOI: 10.1371/journal.pone.0095958] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 04/01/2014] [Indexed: 12/21/2022] Open
Abstract
Prion diseases induce neurodegeneration in specific brain areas for undetermined reasons. A thorough understanding of the localization of the disease-causing molecule, the prion protein (PrP), could inform on this issue but previous studies have generated conflicting conclusions. One of the more intriguing disagreements is whether PrP is synthesized by astrocytes. We developed a knock-in reporter mouse line in which the coding sequence of the PrP expressing gene (Prnp), was replaced with that for green fluorescent protein (GFP). Native GFP fluorescence intensity varied between and within brain regions. GFP was present in astrocytes but did not increase during reactive gliosis induced by scrapie prion infection. Therefore, reactive gliosis associated with prion diseases does not cause an acceleration of local PrP production. In addition to aiding in Prnp gene activity studies, this reporter mouse line will likely prove useful for analysis of chimeric animals produced by stem cell and tissue transplantation experiments.
Collapse
|
41
|
Prion protein misfolding, strains, and neurotoxicity: an update from studies on Mammalian prions. Int J Cell Biol 2013; 2013:910314. [PMID: 24454379 PMCID: PMC3884631 DOI: 10.1155/2013/910314] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 11/10/2013] [Accepted: 11/11/2013] [Indexed: 11/17/2022] Open
Abstract
Prion diseases, also known as transmissible spongiform encephalopathies (TSEs), are a group of fatal neurodegenerative disorders affecting humans and other mammalian species. The central event in TSE pathogenesis is the conformational conversion of the cellular prion protein, PrPC, into the aggregate, β-sheet rich, amyloidogenic form, PrPSc. Increasing evidence indicates that distinct PrPSc conformers, forming distinct ordered aggregates, can encipher the phenotypic TSE variants related to prion strains. Prion strains are TSE isolates that, after inoculation into syngenic hosts, cause disease with distinct characteristics, such as incubation period, pattern of PrPSc distribution, and regional severity of histopathological changes in the brain. In analogy with other amyloid forming proteins, PrPSc toxicity is thought to derive from the existence of various intermediate structures prior to the amyloid fiber formation and/or their specific interaction with membranes. The latter appears particularly relevant for the pathogenesis of TSEs associated with GPI-anchored PrPSc, which involves major cellular membrane distortions in neurons. In this review, we update the current knowledge on the molecular mechanisms underlying three fundamental aspects of the basic biology of prions such as the putative mechanism of prion protein conversion to the pathogenic form PrPSc and its propagation, the molecular basis of prion strains, and the mechanism of induced neurotoxicity by PrPSc aggregates.
Collapse
|