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
|
Benarroch E. What Are the Roles of Cellular Prion Protein in Normal and Pathologic Conditions? Neurology 2024; 102:e209272. [PMID: 38484222 DOI: 10.1212/wnl.0000000000209272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 03/19/2024] Open
|
3
|
Simmons SM, Bartz JC. Strain-Specific Targeting and Destruction of Cells by Prions. BIOLOGY 2024; 13:57. [PMID: 38275733 PMCID: PMC10813089 DOI: 10.3390/biology13010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Prion diseases are caused by the disease-specific self-templating infectious conformation of the host-encoded prion protein, PrPSc. Prion strains are operationally defined as a heritable phenotype of disease under controlled conditions. One of the hallmark phenotypes of prion strain diversity is tropism within and between tissues. A defining feature of prion strains is the regional distribution of PrPSc in the CNS. Additionally, in both natural and experimental prion disease, stark differences in the tropism of prions in secondary lymphoreticular system tissues occur. The mechanism underlying prion tropism is unknown; however, several possible hypotheses have been proposed. Clinical target areas are prion strain-specific populations of neurons within the CNS that are susceptible to neurodegeneration following the replication of prions past a toxic threshold. Alternatively, the switch from a replicative to toxic form of PrPSc may drive prion tropism. The normal form of the prion protein, PrPC, is required for prion formation. More recent evidence suggests that it can mediate prion and prion-like disease neurodegeneration. In vitro systems for prion formation have indicated that cellular cofactors contribute to prion formation. Since these cofactors can be strain specific, this has led to the hypothesis that the distribution of prion formation cofactors can influence prion tropism. Overall, there is evidence to support several mechanisms of prion strain tropism; however, a unified theory has yet to emerge.
Collapse
Affiliation(s)
| | - Jason C. Bartz
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE 68178, USA;
| |
Collapse
|
4
|
Wu J, Wang X, Lakkaraju A, Sternke-Hoffmann R, Qureshi BM, Aguzzi A, Luo J. Channel Activities of the Full-Length Prion and Truncated Proteins. ACS Chem Neurosci 2024; 15:98-107. [PMID: 38096481 DOI: 10.1021/acschemneuro.3c00412] [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: 01/04/2024] Open
Abstract
Prion diseases are fatal neurodegenerative disorders characterized by the conversion of the cellular prion protein (PrPC) into a misfolded prion form, which is believed to disrupt the cellular membranes. However, the exact mechanisms underlying prion toxicity, including the formation of membrane pores, are not fully understood. The prion protein consists of two domains: a globular domain (GD) and a flexible N-terminus (FT) domain. Although a proximal polybasic amino acid (FT(23-31) sequence of FT is a prerequisite for cellular membrane permeabilization, other functional domain regions may modulate its effects. Through single-channel electrical recordings and cryo-electron microscopy (cryo-EM), we discovered that the FT(23-50) fragment forms pore-shaped oligomers and plays a dominant role in membrane permeabilization within the full-length mouse prion protein (mPrP(23-230)). In contrast, the FT(51-110) domain or the C-terminal domain downregulate the channel activity of FT(23-50) and mPrP(23-230). The addition of prion mimetic antibody, POM1 significantly amplifies mPrP(23-230) membrane permeabilization, whereas POM1_Y104A, a mutant that binds to PrP but cannot elicit toxicity, has a negligible effect on membrane permeabilization. Additionally, the anti-N-terminal antibody POM2 or Cu2+ binds to the FT domain, subsequently enhancing the FT(23-110) channel activity. Importantly, our setup provides a novel approach without an external fused protein to examine the channel activity of truncated PrP in the lipid membranes. We therefore propose that the primary N-terminal residues are essential for membrane permeabilization, while other functional segments of PrP play a vital role in modulating the pathological effects of PrP-mediated neurotoxicity.
Collapse
Affiliation(s)
- Jinming Wu
- Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Xue Wang
- Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Asvin Lakkaraju
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | | | - Bilal M Qureshi
- Scientific Center for Optical and Electron Microscopy (ScopeM), ETH Zurich, Zurich 8093, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Jinghui Luo
- Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland
| |
Collapse
|
5
|
Mollica L, Giachin G. Recognition Mechanisms between a Nanobody and Disordered Epitopes of the Human Prion Protein: An Integrative Molecular Dynamics Study. J Chem Inf Model 2022; 63:531-545. [PMID: 36580661 PMCID: PMC9875307 DOI: 10.1021/acs.jcim.2c01062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Immunotherapy using antibodies to target the aggregation of flexible proteins holds promise for therapeutic interventions in neurodegenerative diseases caused by protein misfolding. Prions or PrPSc, the causal agents of transmissible spongiform encephalopathies (TSE), represent a model target for immunotherapies as TSE are prototypical protein misfolding diseases. The X-ray crystal structure of the wild-type (WT) human prion protein (HuPrP) bound to a camelid antibody fragment, denoted as Nanobody 484 (Nb484), has been previously solved. Nb484 was found to inhibit prion aggregation in vitro through a unique mechanism of structural stabilization of two disordered epitopes, that is, the palindromic motif (residues 113-120) and the β2-α2 loop region (residues 164-185). The study of the structural basis for antibody recognition of flexible proteins requires appropriate sampling techniques for the identification of conformational states occurring in disordered epitopes. To elucidate the Nb484-HuPrP recognition mechanisms, here we applied molecular dynamics (MD) simulations complemented with available NMR and X-ray crystallography data collected on the WT HuPrP to describe the conformational spaces occurring on HuPrP prior to Nb484 binding. We observe the experimentally determined binding competent conformations within the ensembles of pre-existing conformational states in solution before binding. We also described the Nb484 recognition mechanisms in two HuPrP carrying a polymorphism (E219K) and a TSE-causing mutation (V210I). Our hybrid approaches allow the identification of dynamic conformational landscapes existing on HuPrP and highly characterized by molecular disorder to identify physiologically relevant and druggable transitions.
Collapse
Affiliation(s)
- Luca Mollica
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, 20090 Milan, Italy,
| | - Gabriele Giachin
- Department
of Chemical Sciences (DiSC), University
of Padua, 35131 Padova, Italy,
| |
Collapse
|
6
|
Muacevic A, Adler JR. Mitogen Activated Protein Kinase (MAPK) Activation, p53, and Autophagy Inhibition Characterize the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Spike Protein Induced Neurotoxicity. Cureus 2022; 14:e32361. [PMID: 36514706 PMCID: PMC9733976 DOI: 10.7759/cureus.32361] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2022] [Indexed: 12/13/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein and prions use common pathogenic pathways to induce toxicity in neurons. Infectious prions rapidly activate the p38 mitogen activated protein kinase (MAPK) pathway, and SARS-CoV-2 spike proteins rapidly activate both the p38 MAPK and c-Jun NH2-terminal kinase (JNK) pathways through toll-like receptor signaling, indicating the potential for similar neurotoxicity, causing prion and prion-like disease. In this review, we analyze the roles of autophagy inhibition, molecular mimicry, elevated intracellular p53 levels and reduced Wild-type p53-induced phosphatase 1 (Wip1) and dual-specificity phosphatase (DUSP) expression in neurons in the disease process. The pathways induced by the spike protein via toll-like receptor activation induce both the upregulation of PrPC (the normal isoform of the prion protein, PrP) and the expression of β amyloid. Through the spike-protein-dependent elevation of p53 levels via β amyloid metabolism, increased PrPC expression can lead to PrP misfolding and impaired autophagy, generating prion disease. We conclude that, according to the age of the spike protein-exposed patient and the state of their cellular autophagy activity, excess sustained activity of p53 in neurons may be a catalytic factor in neurodegeneration. An autoimmune reaction via molecular mimicry likely also contributes to neurological symptoms. Overall results suggest that neurodegeneration is in part due to the intensity and duration of spike protein exposure, patient advanced age, cellular autophagy activity, and activation, function and regulation of p53. Finally, the neurologically damaging effects can be cumulatively spike-protein dependent, whether exposure is by natural infection or, more substantially, by repeated mRNA vaccination.
Collapse
|
7
|
Reimann RR, Puzio M, Rosati A, Emmenegger M, Schneider BL, Valdés P, Huang D, Caflisch A, Aguzzi A. Rapid ex vivo reverse genetics identifies the essential determinants of prion protein toxicity. Brain Pathol 2022; 33:e13130. [PMID: 36329611 PMCID: PMC10041163 DOI: 10.1111/bpa.13130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
The cellular prion protein PrPC mediates the neurotoxicity of prions and other protein aggregates through poorly understood mechanisms. Antibody-derived ligands against the globular domain of PrPC (GDL) can also initiate neurotoxicity by inducing an intramolecular R208 -H140 hydrogen bond ("H-latch") between the α2-α3 and β2-α2 loops of PrPC . Importantly, GDL that suppresses the H-latch prolong the life of prion-infected mice, suggesting that GDL toxicity and prion infections exploit convergent pathways. To define the structural underpinnings of these phenomena, we transduced 19 individual PrPC variants to PrPC -deficient cerebellar organotypic cultured slices using adenovirus-associated viral vectors (AAV). We report that GDL toxicity requires a single N-proximal cationic residue (K27 or R27 ) within PrPC . Alanine substitution of K27 also prevented the toxicity of PrPC mutants that induce Shmerling syndrome, a neurodegenerative disease that is suppressed by co-expression of wild-type PrPC . K27 may represent an actionable target for compounds aimed at preventing prion-related neurodegeneration.
Collapse
Affiliation(s)
| | - Martina Puzio
- Institute of Neuropathology University of Zurich Zurich Switzerland
| | - Antonella Rosati
- Institute of Neuropathology University of Zurich Zurich Switzerland
| | - Marc Emmenegger
- Institute of Neuropathology University of Zurich Zurich Switzerland
| | - Bernard L. Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne Lausanne Switzerland
| | - Pamela Valdés
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne Lausanne Switzerland
| | - Danzhi Huang
- Department of Biochemistry University of Zürich Zürich Switzerland
| | - Amedeo Caflisch
- Department of Biochemistry University of Zürich Zürich Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology University of Zurich Zurich Switzerland
| |
Collapse
|
8
|
Mercer RCC, Harris DA. Mechanisms of prion-induced toxicity. Cell Tissue Res 2022; 392:81-96. [PMID: 36070155 DOI: 10.1007/s00441-022-03683-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022]
Abstract
Prion diseases are devastating neurodegenerative diseases caused by the structural conversion of the normally benign prion protein (PrPC) to an infectious, disease-associated, conformer, PrPSc. After decades of intense research, much is known about the self-templated prion conversion process, a phenomenon which is now understood to be operative in other more common neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. In this review, we provide the current state of knowledge concerning a relatively poorly understood aspect of prion diseases: mechanisms of neurotoxicity. We provide an overview of proposed functions of PrPC and its interactions with other extracellular proteins in the central nervous system, in vivo and in vitro models used to delineate signaling events downstream of prion propagation, the application of omics technologies, and the emerging appreciation of the role played by non-neuronal cell types in pathogenesis.
Collapse
Affiliation(s)
- Robert C C Mercer
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
9
|
A conformational switch controlling the toxicity of the prion protein. Nat Struct Mol Biol 2022; 29:831-840. [PMID: 35948768 PMCID: PMC9371974 DOI: 10.1038/s41594-022-00814-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 07/06/2022] [Indexed: 11/08/2022]
Abstract
Prion infections cause conformational changes of the cellular prion protein (PrPC) and lead to progressive neurological impairment. Here we show that toxic, prion-mimetic ligands induce an intramolecular R208-H140 hydrogen bond ('H-latch'), altering the flexibility of the α2-α3 and β2-α2 loops of PrPC. Expression of a PrP2Cys mutant mimicking the H-latch was constitutively toxic, whereas a PrPR207A mutant unable to form the H-latch conferred resistance to prion infection. High-affinity ligands that prevented H-latch induction repressed prion-related neurodegeneration in organotypic cerebellar cultures. We then selected phage-displayed ligands binding wild-type PrPC, but not PrP2Cys. These binders depopulated H-latched conformers and conferred protection against prion toxicity. Finally, brain-specific expression of an antibody rationally designed to prevent H-latch formation prolonged the life of prion-infected mice despite unhampered prion propagation, confirming that the H-latch is an important reporter of prion neurotoxicity.
Collapse
|
10
|
Requena JR. Unlatching a window into the molecular landscape of prion toxicity. Nat Struct Mol Biol 2022; 29:733-735. [PMID: 35948769 DOI: 10.1038/s41594-022-00817-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jesús R Requena
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain.
| |
Collapse
|
11
|
Ilie IM, Bacci M, Vitalis A, Caflisch A. Antibody binding modulates the dynamics of the membrane-bound prion protein. Biophys J 2022; 121:2813-2825. [PMID: 35672948 PMCID: PMC9382331 DOI: 10.1016/j.bpj.2022.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/20/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Abstract
Misfolding of the cellular prion protein (PrPC) is associated with lethal neurodegeneration. PrPC consists of a flexible tail (residues 23-123) and a globular domain (residues 124-231) whose C-terminal end is anchored to the cell membrane. The neurotoxic antibody POM1 and the innocuous antibody POM6 recognize the globular domain. Experimental evidence indicates that POM1 binding to PrPC emulates the influence on PrPC of the misfolded prion protein (PrPSc) while the binding of POM6 has the opposite biological response. Little is known about the potential interactions between flexible tail, globular domain, and the membrane. Here, we used atomistic simulations to investigate how these interactions are modulated by the binding of the Fab fragments of POM1 and POM6 to PrPC and by interstitial sequence truncations to the flexible tail. The simulations show that the binding of the antibodies restricts the range of orientations of the globular domain with respect to the membrane and decreases the distance between tail and membrane. Five of the six sequence truncations influence only marginally this distance and the contact patterns between tail and globular domain. The only exception is a truncation coupled to a charge inversion mutation of four N-terminal residues, which increases the distance of the flexible tail from the membrane. The interactions of the flexible tail and globular domain are modulated differently by the two antibodies.
Collapse
Affiliation(s)
- Ioana M Ilie
- Department of Biochemistry, University of Zürich, Zürich, Switzerland
| | - Marco Bacci
- Department of Biochemistry, University of Zürich, Zürich, Switzerland
| | - Andreas Vitalis
- Department of Biochemistry, University of Zürich, Zürich, Switzerland
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
12
|
Abstract
Amyloids are protein aggregates bearing a highly ordered cross β structural motif, which may be functional but are mostly pathogenic. Their formation, deposition in tissues and consequent organ dysfunction is the central event in amyloidogenic diseases. Such protein aggregation may be brought about by conformational changes, and much attention has been directed toward factors like metal binding, post-translational modifications, mutations of protein etc., which eventually affect the reactivity and cytotoxicity of the associated proteins. Over the past decade, a global effort from different groups working on these misfolded/unfolded proteins/peptides has revealed that the amino acid residues in the second coordination sphere of the active sites of amyloidogenic proteins/peptides cause changes in H-bonding pattern or protein-protein interactions, which dramatically alter the structure and reactivity of these proteins/peptides. These second sphere effects not only determine the binding of transition metals and cofactors, which define the pathology of some of these diseases, but also change the mechanism of redox reactions catalyzed by these proteins/peptides and form the basis of oxidative damage associated with these amyloidogenic diseases. The present review seeks to discuss such second sphere modifications and their ramifications in the etiopathology of some representative amyloidogenic diseases like Alzheimer's disease (AD), type 2 diabetes mellitus (T2Dm), Parkinson's disease (PD), Huntington's disease (HD), and prion diseases.
Collapse
Affiliation(s)
- Madhuparna Roy
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Arnab Kumar Nath
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
13
|
Khadka A, Spiers JG, Cheng L, Hill AF. Extracellular vesicles with diagnostic and therapeutic potential for prion diseases. Cell Tissue Res 2022; 392:247-267. [PMID: 35394216 PMCID: PMC10113352 DOI: 10.1007/s00441-022-03621-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/25/2022] [Indexed: 12/14/2022]
Abstract
Prion diseases (PrD) or transmissible spongiform encephalopathies (TSE) are invariably fatal and pathogenic neurodegenerative disorders caused by the self-propagated misfolding of cellular prion protein (PrPC) to the neurotoxic pathogenic form (PrPTSE) via a yet undefined but profoundly complex mechanism. Despite several decades of research on PrD, the basic understanding of where and how PrPC is transformed to the misfolded, aggregation-prone and pathogenic PrPTSE remains elusive. The primary clinical hallmarks of PrD include vacuolation-associated spongiform changes and PrPTSE accumulation in neural tissue together with astrogliosis. The difficulty in unravelling the disease mechanisms has been related to the rare occurrence and long incubation period (over decades) followed by a very short clinical phase (few months). Additional challenge in unravelling the disease is implicated to the unique nature of the agent, its complexity and strain diversity, resulting in the heterogeneity of the clinical manifestations and potentially diverse disease mechanisms. Recent advances in tissue isolation and processing techniques have identified novel means of intercellular communication through extracellular vesicles (EVs) that contribute to PrPTSE transmission in PrD. This review will comprehensively discuss PrPTSE transmission and neurotoxicity, focusing on the role of EVs in disease progression, biomarker discovery and potential therapeutic agents for the treatment of PrD.
Collapse
Affiliation(s)
- Arun Khadka
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Jereme G Spiers
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Lesley Cheng
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Andrew F Hill
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia. .,Institute for Health and Sport, Victoria University, Footscray, VIC, Australia.
| |
Collapse
|
14
|
The cellular prion protein interacts with and promotes the activity of Na,K-ATPases. PLoS One 2021; 16:e0258682. [PMID: 34847154 PMCID: PMC8631662 DOI: 10.1371/journal.pone.0258682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 10/02/2021] [Indexed: 12/23/2022] Open
Abstract
The prion protein (PrP) is best known for its ability to cause fatal neurodegenerative diseases in humans and animals. Here, we revisited its molecular environment in the brain using a well-developed affinity-capture mass spectrometry workflow that offers robust relative quantitation. The analysis confirmed many previously reported interactions. It also pointed toward a profound enrichment of Na,K-ATPases (NKAs) in proximity to cellular PrP (PrPC). Follow-on work validated the interaction, demonstrated partial co-localization of the ATP1A1 and PrPC, and revealed that cells exposed to cardiac glycoside (CG) inhibitors of NKAs exhibit correlated changes to the steady-state levels of both proteins. Moreover, the presence of PrPC was observed to promote the ion uptake activity of NKAs in a human co-culture paradigm of differentiated neurons and glia cells, and in mouse neuroblastoma cells. Consistent with this finding, changes in the expression of 5’-nucleotidase that manifest in wild-type cells in response to CG exposure can also be observed in untreated PrPC-deficient cells. Finally, the endoproteolytic cleavage of the glial fibrillary acidic protein, a hallmark of late-stage prion disease, can also be induced by CGs, raising the prospect that a loss of NKA activity may contribute to the pathobiology of prion diseases.
Collapse
|
15
|
Linsenmeier L, Mohammadi B, Shafiq M, Frontzek K, Bär J, Shrivastava AN, Damme M, Song F, Schwarz A, Da Vela S, Massignan T, Jung S, Correia A, Schmitz M, Puig B, Hornemann S, Zerr I, Tatzelt J, Biasini E, Saftig P, Schweizer M, Svergun D, Amin L, Mazzola F, Varani L, Thapa S, Gilch S, Schätzl H, Harris DA, Triller A, Mikhaylova M, Aguzzi A, Altmeppen HC, Glatzel M. Ligands binding to the prion protein induce its proteolytic release with therapeutic potential in neurodegenerative proteinopathies. SCIENCE ADVANCES 2021; 7:eabj1826. [PMID: 34818048 PMCID: PMC8612689 DOI: 10.1126/sciadv.abj1826] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/20/2021] [Indexed: 05/07/2023]
Abstract
The prion protein (PrPC) is a central player in neurodegenerative diseases, such as prion diseases or Alzheimer’s disease. In contrast to disease-promoting cell surface PrPC, extracellular fragments act neuroprotective by blocking neurotoxic disease-associated protein conformers. Fittingly, PrPC release by the metalloprotease ADAM10 represents a protective mechanism. We used biochemical, cell biological, morphological, and structural methods to investigate mechanisms stimulating this proteolytic shedding. Shed PrP negatively correlates with prion conversion and is markedly redistributed in murine brain in the presence of prion deposits or amyloid plaques, indicating a sequestrating activity. PrP-directed ligands cause structural changes in PrPC and increased shedding in cells and organotypic brain slice cultures. As an exception, some PrP-directed antibodies targeting repetitive epitopes do not cause shedding but surface clustering, endocytosis, and degradation of PrPC. Both mechanisms may contribute to beneficial actions described for PrP-directed ligands and pave the way for new therapeutic strategies against currently incurable neurodegenerative diseases.
Collapse
Affiliation(s)
- Luise Linsenmeier
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Karl Frontzek
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Julia Bär
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Molecular Neurobiology Hamburg (ZMNH), UKE, Hamburg, Germany
| | - Amulya N. Shrivastava
- École Normale Supérieure, Institut de Biologie de l’ENS (IBENS), INSERM, CNRS, PSL Research University, Paris, France
| | - Markus Damme
- Institute of Biochemistry, Christian Albrechts University, Kiel, Germany
| | - Feizhi Song
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Alexander Schwarz
- Institute of Nanostructure and Solid State Physics, Universität Hamburg, Hamburg, Germany
| | - Stefano Da Vela
- European Molecular Biology Laboratory (EMBL), Hamburg, Germany
| | - Tania Massignan
- Dulbecco Telethon Laboratory of Prions and Amyloids, CIBIO, University of Trento, Trento, Italy
| | - Sebastian Jung
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Angela Correia
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Matthias Schmitz
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation, UKE, Hamburg, Germany
| | - Simone Hornemann
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jörg Tatzelt
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Cluster of Excellence RESOLV, Bochum, Germany
| | - Emiliano Biasini
- Dulbecco Telethon Laboratory of Prions and Amyloids, CIBIO, University of Trento, Trento, Italy
| | - Paul Saftig
- Institute of Biochemistry, Christian Albrechts University, Kiel, Germany
| | | | - Dmitri Svergun
- European Molecular Biology Laboratory (EMBL), Hamburg, Germany
| | - Ladan Amin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Federica Mazzola
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Luca Varani
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Simrika Thapa
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, Canada
| | - Sabine Gilch
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, Canada
| | - Hermann Schätzl
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, Canada
| | - David A. Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Antoine Triller
- École Normale Supérieure, Institut de Biologie de l’ENS (IBENS), INSERM, CNRS, PSL Research University, Paris, France
| | - Marina Mikhaylova
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Molecular Neurobiology Hamburg (ZMNH), UKE, Hamburg, Germany
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Hermann C. Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
16
|
Zhang X, Pan YH, Chen Y, Pan C, Ma J, Yuan C, Yu G, Ma J. The protease-sensitive N-terminal polybasic region of prion protein modulates its conversion to the pathogenic prion conformer. J Biol Chem 2021; 297:101344. [PMID: 34710372 PMCID: PMC8604679 DOI: 10.1016/j.jbc.2021.101344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 11/17/2022] Open
Abstract
Conversion of normal prion protein (PrPC) to the pathogenic PrPSc conformer is central to prion diseases such as Creutzfeldt-Jakob disease and scrapie; however, the detailed mechanism of this conversion remains obscure. To investigate how the N-terminal polybasic region of PrP (NPR) influences the PrPC-to-PrPSc conversion, we analyzed two PrP mutants: ΔN6 (deletion of all six amino acids in NPR) and Met4-1 (replacement of four positively charged amino acids in NPR with methionine). We found that ΔN6 and Met4-1 differentially impacted the binding of recombinant PrP (recPrP) to the negatively charged phospholipid 1-palmitoyl-2-oleoylphosphatidylglycerol, a nonprotein cofactor that facilitates PrP conversion. Both mutant recPrPs were able to form recombinant prion (recPrPSc) in vitro, but the convertibility was greatly reduced, with ΔN6 displaying the lowest convertibility. Prion infection assays in mammalian RK13 cells expressing WT or NPR-mutant PrPs confirmed these differences in convertibility, indicating that the NPR affects the conversion of both bacterially expressed recPrP and post-translationally modified PrP in eukaryotic cells. We also found that both WT and mutant recPrPSc conformers caused prion disease in WT mice with a 100% attack rate, but the incubation times and neuropathological changes caused by two recPrPSc mutants were significantly different from each other and from that of WT recPrPSc. Together, our results support that the NPR greatly influences PrPC-to-PrPSc conversion, but it is not essential for the generation of PrPSc. Moreover, the significant differences between ΔN6 and Met4-1 suggest that not only charge but also the identity of amino acids in NPR is important to PrP conversion.
Collapse
Affiliation(s)
- Xiangyi Zhang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China
| | - Yi-Hsuan Pan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China
| | - Ying Chen
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China
| | - Chenhua Pan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China
| | - Ji Ma
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China
| | - Chonggang Yuan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China
| | - Guohua Yu
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, School of Life Sciences, Longyan University, Longyan, China
| | - Jiyan Ma
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China; Department of Neurodegeneraive Science, Van Andel Institute, Grand Rapids, Michigan, USA; Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
17
|
Adhikari UK, Tayebi M. Epitope-specific anti-PrP antibody toxicity: a comparative in-silico study of human and mouse prion proteins. Prion 2021; 15:155-176. [PMID: 34632945 PMCID: PMC8900626 DOI: 10.1080/19336896.2021.1964326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Despite having therapeutic potential, anti-PrP antibodies caused a major controversy due to their neurotoxic effects. For instance, treating mice with ICSM antibodies delayed prion disease onset, but both were found to be either toxic or innocuous to neurons by researchers following cross-linking PrPC. In order to elucidate and understand the reasons that led to these contradictory outcomes, we conducted a comprehensive in silico study to assess the antibody-specific toxicity. Since most therapeutic anti-PrP antibodies were generated against human truncated recombinant PrP91-231 or full-length mouse PrP23-231, we reasoned that host specificity (human vs murine) of PrPC might influence the nature of the specific epitopes recognized by these antibodies at the structural level possibly explaining the 'toxicity' discrepancies reported previously. Initially, molecular dynamics simulation and pro-motif analysis of full-length human (hu)PrP and mouse (mo)PrP 3D structure displayed conspicuous structural differences between huPrP and moPrP. We identified 10 huPrP and 6 moPrP linear B-cell epitopes from the prion protein 3D structure where 5 out of 10 huPrP and 3 out of 6 moPrP B-cell epitopes were predicted to be potentially toxic in immunoinformatics approaches. Herein, we demonstrate that some of the predicted potentially 'toxic' epitopes identified by the in silico analysis were similar to the epitopes recognized by the toxic antibodies such as ICSM18 (146-159), POM1 (138-147), D18 (133-157), ICSM35 (91-110), D13 (95-103) and POM3 (95-100). This in silico study reveals the role of host specificity of PrPC in epitope-specific anti-PrP antibody toxicity.
Collapse
Affiliation(s)
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
18
|
Calcineurin Activation by Prion Protein Induces Neurotoxicity via Mitochondrial Reactive Oxygen Species. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5572129. [PMID: 34394828 PMCID: PMC8363446 DOI: 10.1155/2021/5572129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/01/2021] [Accepted: 07/14/2021] [Indexed: 01/02/2023]
Abstract
Prion diseases are caused by PrPsc accumulation in the brain, which triggers dysfunctional mitochondrial injury and reactive oxygen species (ROS) generation in neurons. Recent studies on prion diseases suggest that endoplasmic reticulum (ER) stress induced by misfolding proteins such as misfolded prion protein results in activation of calcineurin. Calcineurin is a calcium-related protein phosphatase of type 2B that exists in copious quantities in the brain and acts as a critical nodal component in the control of cellular functions. To investigate the relationship between calcineurin and intracellular ROS, we assessed the alteration of CaN and ROS induced by prion peptide (PrP) 106-126. Human prion peptide increased mitochondrial ROS by activating calcineurin, and the inhibition of calcineurin activity protected mitochondrial function and neuronal apoptosis in neuronal cells. These results suggest that calcineurin plays a pivotal role in neuronal apoptosis by mediating mitochondrial injury and ROS in prion diseases.
Collapse
|
19
|
Lakkaraju AKK, Frontzek K, Lemes E, Herrmann U, Losa M, Marpakwar R, Aguzzi A. Loss of PIKfyve drives the spongiform degeneration in prion diseases. EMBO Mol Med 2021; 13:e14714. [PMID: 34291577 PMCID: PMC8518562 DOI: 10.15252/emmm.202114714] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 11/21/2022] Open
Abstract
Brain‐matter vacuolation is a defining trait of all prion diseases, yet its cause is unknown. Here, we report that prion infection and prion‐mimetic antibodies deplete the phosphoinositide kinase PIKfyve—which controls endolysosomal maturation—from mouse brains, cultured cells, organotypic brain slices, and brains of Creutzfeldt‐Jakob disease victims. We found that PIKfyve is acylated by the acyltransferases zDHHC9 and zDHHC21, whose juxtavesicular topology is disturbed by prion infection, resulting in PIKfyve deacylation and rapid degradation, as well as endolysosomal hypertrophy and activation of TFEB‐dependent lysosomal enzymes. A protracted unfolded protein response (UPR), typical of prion diseases, also induced PIKfyve deacylation and degradation. Conversely, UPR antagonists restored PIKfyve levels in prion‐infected cells. Overexpression of zDHHC9 and zDHHC21, administration of the antiprion polythiophene LIN5044, or supplementation with the PIKfyve reaction product PI(3,5)P2 suppressed prion‐induced vacuolation and restored lysosomal homeostasis. Thus, PIKfyve emerges as a central mediator of vacuolation and neurotoxicity in prion diseases.
Collapse
Affiliation(s)
| | - Karl Frontzek
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Emina Lemes
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Uli Herrmann
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Marco Losa
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | | | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| |
Collapse
|
20
|
Moon JH, Hong JM, Park SY. The antidiabetic drug troglitazone protects against PrP (106‑126)‑induced neurotoxicity via the PPARγ‑autophagy pathway in neuronal cells. Mol Med Rep 2021; 23:430. [PMID: 33846779 PMCID: PMC8047904 DOI: 10.3892/mmr.2021.12069] [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: 09/04/2020] [Accepted: 03/16/2021] [Indexed: 12/11/2022] Open
Abstract
Prion diseases, which involve the alteration of cellular prion protein into a misfolded isoform, disrupt the central nervous systems of humans and animals alike. Prior research has suggested that peroxisome proliferator-activator receptor (PPAR)γ and autophagy provide some protection against neurodegeneration. PPARs are critical to lipid metabolism regulation and autophagy is one of the main cellular mechanisms by which cell function and homeostasis is maintained. The present study examined the effect of troglitazone, a PPARγ agonist, on autophagy flux in a prion peptide (PrP) (106–126)-mediated neurodegeneration model. Western blot analysis confirmed that treatment with troglitazone increased LC3-II and p62 protein expression, whereas an excessive increase in autophagosomes was verified by transmission electron microscopy. Troglitazone weakened PrP (106–126)-mediated neurotoxicity via PPARγ activation and autophagy flux inhibition. A PPARγ antagonist blocked PPARγ activation as well as the neuroprotective effects induced by troglitazone treatment, indicating that PPARγ deactivation impaired troglitazone-mediated protective effects. In conclusion, the present study demonstrated that troglitazone protected primary neuronal cells against PrP (106–126)-induced neuronal cell death by inhibiting autophagic flux and activating PPARγ signals. These results suggested that troglitazone may be a useful therapeutic agent for the treatment of neurodegenerative disorders and prion diseases.
Collapse
Affiliation(s)
- Ji-Hong Moon
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| | - Jeong-Min Hong
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| |
Collapse
|
21
|
Neuroinflammation in Prion Disease. Int J Mol Sci 2021; 22:ijms22042196. [PMID: 33672129 PMCID: PMC7926464 DOI: 10.3390/ijms22042196] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/20/2021] [Accepted: 02/20/2021] [Indexed: 12/24/2022] Open
Abstract
Neuroinflammation, typically manifest as microglial activation and astrogliosis accompanied by transcriptomic alterations, represents a common hallmark of various neurodegenerative conditions including prion diseases. Microglia play an overall neuroprotective role in prion disease, whereas reactive astrocytes with aberrant phenotypes propagate prions and contribute to prion-induced neurodegeneration. The existence of heterogeneous subpopulations and dual functions of microglia and astrocytes in prion disease make them potential targets for therapeutic intervention. A variety of neuroinflammation-related molecules are involved in prion pathogenesis. Therapeutics targeting neuroinflammation represents a novel approach to combat prion disease. Deciphering neuroinflammation in prion disease will deepen our understanding of pathogenesis of other neurodegenerative disorders.
Collapse
|
22
|
Roux A, Wang X, Becker K, Ma J. Modeling α-Synucleinopathy in Organotypic Brain Slice Culture with Preformed α-Synuclein Amyloid Fibrils. JOURNAL OF PARKINSONS DISEASE 2020; 10:1397-1410. [PMID: 32716318 PMCID: PMC7683096 DOI: 10.3233/jpd-202026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Synucleinopathy is a group of neurodegenerative disorders characterized by neurodegeneration and accumulation of alpha-synuclein (α-syn) aggregates in various brain regions. The detailed mechanism of α-syn-caused neurotoxicity remains obscure, which is partly due to the lack of a suitable model that retains the in vivo three-dimensional cellular network and allows a convenient dissection of the neurotoxic pathways. Recent studies revealed that the pre-formed recombinant α-syn amyloid fibrils (PFFs) induce a robust accumulation of pathogenic α-syn species in cultured cells and animals. Objective: Our goal is to determine whether PFFs are able to induce the pathogenic α-syn accumulation and neurotoxicity in organotypic brain slice culture, an ex vivo system that retains the in vivo three-dimensional cell-cell connections. Methods/Results: Adding PFFs to cultured wild-type rat or mouse brain slices induced a time-dependent accumulation of pathogenic α-syn species, which was indicated by α-syn phosphorylated at serine 129 (pα-syn). The PFF-induced pα-syn was abolished in brain slices prepared from α-syn null mice, suggesting that the pα-syn is from the phosphorylation of endogenous α-syn. Human PFFs also induced pα-syn in brain slices prepared from mice expressing human α-syn on a mouse α-syn-null background. Furthermore, the synaptophysin immunoreactivity was inversely associated with pα-syn accumulation and an increase of neuronal loss was detected. Conclusion: PFF-treatment of brain slices is able to induce key pathological features of synucleinopathy: pα-syn accumulation and neurotoxicity. This model will be useful for investigating the neurotoxic mechanism and evaluating efficacy of therapeutic approaches.
Collapse
Affiliation(s)
- Amandine Roux
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Xinhe Wang
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Katelyn Becker
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Jiyan Ma
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| |
Collapse
|
23
|
Recent developments in antibody therapeutics against prion disease. Emerg Top Life Sci 2020; 4:169-173. [PMID: 32633322 DOI: 10.1042/etls20200002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022]
Abstract
Preclinical evidence indicates that prion diseases can respond favorably to passive immunotherapy. However, certain antibodies to the cellular prion protein PrPC can be toxic. Comprehensive studies of structure-function relationships have revealed that the flexible amino-terminal tail of PrPC is instrumental for mediating prion toxicity. In a first-in-human study, an anti-prion antibody has been recently administered to patients diagnosed with sporadic Creutzfeldt-Jakob's disease, the most prevalent human prion disease. Moreover, large-scale serosurveys have mapped the prevalence of naturally occurring human anti-prion autoantibodies in health and disease. Here, we provide a perspective on the limitations and opportunities of therapeutic anti-prion antibodies.
Collapse
|
24
|
Senatore A, Frontzek K, Emmenegger M, Chincisan A, Losa M, Reimann R, Horny G, Guo J, Fels S, Sorce S, Zhu C, George N, Ewert S, Pietzonka T, Hornemann S, Aguzzi A. Protective anti-prion antibodies in human immunoglobulin repertoires. EMBO Mol Med 2020; 12:e12739. [PMID: 32776637 PMCID: PMC7506995 DOI: 10.15252/emmm.202012739] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 01/09/2023] Open
Abstract
Prion immunotherapy may hold great potential, but antibodies against certain PrP epitopes can be neurotoxic. Here, we identified > 6,000 PrP-binding antibodies in a synthetic human Fab phage display library, 49 of which we characterized in detail. Antibodies directed against the flexible tail of PrP conferred neuroprotection against infectious prions. We then mined published repertoires of circulating B cells from healthy humans and found antibodies similar to the protective phage-derived antibodies. When expressed recombinantly, these antibodies exhibited anti-PrP reactivity. Furthermore, we surveyed 48,718 samples from 37,894 hospital patients for the presence of anti-PrP IgGs and found 21 high-titer individuals. The clinical files of these individuals did not reveal any enrichment of specific pathologies, suggesting that anti-PrP autoimmunity is innocuous. The existence of anti-prion antibodies in unbiased human immunological repertoires suggests that they might clear nascent prions early in life. Combined with the reported lack of such antibodies in carriers of disease-associated PRNP mutations, this suggests a link to the low incidence of spontaneous prion diseases in human populations.
Collapse
Affiliation(s)
- Assunta Senatore
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Karl Frontzek
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Marc Emmenegger
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Andra Chincisan
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Marco Losa
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Regina Reimann
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Geraldine Horny
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Jingjing Guo
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Sylvie Fels
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Silvia Sorce
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Caihong Zhu
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Nathalie George
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Stefan Ewert
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Simone Hornemann
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
25
|
POSCAbilities: The Application of the Prion Organotypic Slice Culture Assay to Neurodegenerative Disease Research. Biomolecules 2020; 10:biom10071079. [PMID: 32698402 PMCID: PMC7407827 DOI: 10.3390/biom10071079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/06/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
Prion diseases are fatal, transmissible neurodegenerative disorders whose pathogenesis is driven by the misfolding, self-templating and cell-to-cell spread of the prion protein. Other neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis and Huntington’s disease, share some of these prion-like features, with different aggregation-prone proteins. Consequently, researchers have begun to apply prion-specific techniques, like the prion organotypic slice culture assay (POSCA), to these disorders. In this review we explore the ways in which the prion phenomenon has been used in organotypic cultures to study neurodegenerative diseases from the perspective of protein aggregation and spreading, strain propagation, the role of glia in pathogenesis, and efficacy of drug treatments. We also present an overview of the advantages and disadvantages of this culture system compared to in vivo and in vitro models and provide suggestions for new directions.
Collapse
|
26
|
Rana SVS. Endoplasmic Reticulum Stress Induced by Toxic Elements-a Review of Recent Developments. Biol Trace Elem Res 2020; 196:10-19. [PMID: 31686395 DOI: 10.1007/s12011-019-01903-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022]
Abstract
Endoplasmic reticulum of all eukaryotic cells is a membrane-bound organelle. Under electron microscope it appears as parallel arrays of "rough membranes" and a maze of "smooth vesicles" respectively. It performs various functions in cell, i.e., synthesis of proteins to degradation of xenobiotics. Bioaccumulation of drugs/chemicals/xenobiotics in the cytosol can trigger ER stress. It is recognized by the accumulation of unfolded or misfolded proteins in the lumen of ER. Present review summarizes the present status of knowledge on ER stress caused by toxic elements, viz arsenic, cadmium, lead, mercury, copper, chromium, and nickel. While inorganic arsenic may induce various glucose-related proteins, i.e., GRP78, GRP94 and CHOP, XBP1, and calpains, cadmium upregulates GRP78. Antioxidants like ascorbic acid, NAC, and Se inhibit the expression of UPR. Exposure to lead also changes ER stress related genes, i.e., GRP 78, GRP 94, ATF4, and ATF6. Mercury too upregulates these genes. Nickel, a carcinogenic element upregulates the expression of Bak, cytochrome C, caspase-3, caspase-9, caspase-12, and GADD 153. Much is not known on ER stress caused by nanoparticles. The review describes inter-organelle association between mitochondria and ER. It also discusses the interdependence between oxidative stress and ER stress. A cross talk amongst different cellular components appears essential to disturb pathways leading to cell death. However, these molecular switches within the signaling network used by toxic elements need to be identified. Nevertheless, ER stress especially caused by toxic elements still remains to be an engaging issue.
Collapse
Affiliation(s)
- S V S Rana
- Department of Toxicology, Ch. Charan Singh University, Meerut, 250 004, India.
| |
Collapse
|
27
|
Roseman GP, Wu B, Wadolkowski MA, Harris DA, Millhauser GL. Intrinsic toxicity of the cellular prion protein is regulated by its conserved central region. FASEB J 2020; 34:8734-8748. [PMID: 32385908 DOI: 10.1096/fj.201902749rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/08/2020] [Accepted: 04/20/2020] [Indexed: 11/11/2022]
Abstract
The conserved central region (CR) of PrPC has been hypothesized to serve as a passive linker connecting the protein's toxic N-terminal and globular C-terminal domains. Yet, deletion of the CR causes neonatal fatality in mice, implying the CR possesses a protective function. The CR encompasses the regulatory α-cleavage locus, and additionally facilitates a regulatory metal ion-promoted interaction between the PrPC N- and C-terminal domains. To elucidate the role of the CR and determine why CR deletion generates toxicity, we designed PrPC constructs wherein either the cis-interaction or α-cleavage are selectively prevented. These constructs were interrogated using nuclear magnetic resonance, electrophysiology, and cell viability assays. Our results demonstrate the CR is not a passive linker and the native sequence is crucial for its protective role over the toxic N-terminus, irrespective of α-cleavage or the cis-interaction. Additionally, we find that the CR facilitates homodimerization of PrPC , attenuating the toxicity of the N-terminus.
Collapse
Affiliation(s)
- Graham P Roseman
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Bei Wu
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Mark A Wadolkowski
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Glenn L Millhauser
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| |
Collapse
|
28
|
Kondru N, Manne S, Kokemuller R, Greenlee J, Greenlee MHW, Nichols T, Kong Q, Anantharam V, Kanthasamy A, Halbur P, Kanthasamy AG. An Ex Vivo Brain Slice Culture Model of Chronic Wasting Disease: Implications for Disease Pathogenesis and Therapeutic Development. Sci Rep 2020; 10:7640. [PMID: 32376941 PMCID: PMC7203233 DOI: 10.1038/s41598-020-64456-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 04/12/2020] [Indexed: 12/03/2022] Open
Abstract
Chronic wasting disease (CWD) is a rapidly spreading prion disease of cervids, yet antemortem diagnosis, treatment, and control remain elusive. We recently developed an organotypic slice culture assay for sensitive detection of scrapie prions using ultrasensitive prion seeding. However, this model was not established for CWD prions due to their strong transmission barrier from deer (Odocoileus spp) to standard laboratory mice (Mus musculus). Therefore, we developed and characterized the ex vivo brain slice culture model for CWD, using a transgenic mouse model (Tg12) that expresses the elk (Cervus canadensis) prion protein gene (PRNP). We tested for CWD infectivity in cultured slices using sensitive seeding assays such as real-time quaking-induced conversion (RT-QuIC) and protein misfolding cyclic amplification (PMCA). Slice cultures from Tg12, but not from prnp-/- mice, tested positive for CWD. Slice-generated CWD prions transmitted efficiently to Tg12 mice. Furthermore, we determined the activity of anti-prion compounds and optimized a screening protocol for the infectivity of biological samples in this CWD slice culture model. Our results demonstrate that this integrated brain slice model of CWD enables the study of pathogenic mechanisms with translational implications for controlling CWD.
Collapse
Affiliation(s)
- Naveen Kondru
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Sireesha Manne
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Robyn Kokemuller
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Justin Greenlee
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - M Heather West Greenlee
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Tracy Nichols
- Surveillance, Preparedness and Response Services, Veterinary Services, United States Department of Agriculture, Fort Collins, CO, USA
| | - Qingzhong Kong
- Departments of Pathology and Neurology, Case Western Reserve University, Cleveland, OH, USA
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Patrick Halbur
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.
| |
Collapse
|
29
|
Flønes IH, Ricken G, Klotz S, Lang A, Ströbel T, Dölle C, Kovacs GG, Tzoulis C. Mitochondrial respiratory chain deficiency correlates with the severity of neuropathology in sporadic Creutzfeldt-Jakob disease. Acta Neuropathol Commun 2020; 8:50. [PMID: 32299489 PMCID: PMC7160955 DOI: 10.1186/s40478-020-00915-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/13/2020] [Indexed: 01/30/2023] Open
Abstract
Mitochondrial dysfunction has been implicated in multiple neurodegenerative diseases but remains largely unexplored in Creutzfeldt-Jakob disease. Here, we characterize the mitochondrial respiratory chain at the individual neuron level in the MM1 and VV2 common molecular subtypes of sporadic Creutzfeldt-Jakob disease. Moreover, we investigate the associations between the mitochondrial respiratory chain and neuropathological markers of the disease.Brain tissue from individuals with sporadic Creutzfeldt-Jakob disease and age-matched controls were obtained from the brain collection of the Austrian Creutzfeldt-Jakob Surveillance. The mitochondrial respiratory chain was studied through a dichotomous approach of immunoreactivities in the temporal cortex and the hippocampal subregions of CA4 and CA3.We show that profound deficiency of all mitochondrial respiratory complexes (I-V) occurs in neurons of the severely affected temporal cortex of patients with Creutzfeldt-Jakob disease. This deficiency correlates strongly with the severity of neuropathological changes, including vacuolation of the neuropil, gliosis and disease associated prion protein load. Respiratory chain deficiency is less pronounced in hippocampal CA4 and CA3 regions compared to the temporal cortex. In both areas respiratory chain deficiency shows a predilection for the MM1 molecular subtype of Creutzfeldt-Jakob disease.Our findings indicate that aberrant mitochondrial respiration could be involved early in the pathogenesis of sporadic Creutzfeldt-Jakob disease and contributes to neuronal death, most likely via ATP depletion. Based on these results, we propose that the restricted MRI diffusion profile seen in the brain of patients with sporadic Creutzfeldt-Jakob disease might reflect cytotoxic changes due to neuronal respiratory chain failure and ATP loss.
Collapse
Affiliation(s)
- Irene H Flønes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Gerda Ricken
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Sigrid Klotz
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Alexandra Lang
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Ströbel
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Christian Dölle
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, Vienna, Austria.
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada.
- Laboratory Medicine Program, University Health Network, Toronto, Canada.
| | - Charalampos Tzoulis
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway.
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway.
| |
Collapse
|
30
|
Ma Y, Ma J. Immunotherapy against Prion Disease. Pathogens 2020; 9:E216. [PMID: 32183309 PMCID: PMC7157205 DOI: 10.3390/pathogens9030216] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/12/2020] [Accepted: 03/12/2020] [Indexed: 11/17/2022] Open
Abstract
The term "prion disease" encompasses a group of neurodegenerative diseases affecting both humans and animals. Currently, there is no effective therapy and all forms of prion disease are invariably fatal. Because of (a) the outbreak of bovine spongiform encephalopathy in cattle and variant Creutzfeldt-Jakob disease in humans; (b) the heated debate about the prion hypothesis; and (c) the availability of a natural prion disease in rodents, the understanding of the pathogenic process in prion disease is much more advanced compared to that of other neurodegenerative disorders, which inspired many attempts to develop therapeutic strategies against these fatal diseases. In this review, we focus on immunotherapy against prion disease. We explain our rationale for immunotherapy as a plausible therapeutic choice, review previous trials using either active or passive immunization, and discuss potential strategies for overcoming the hurdles in developing a successful immunotherapy. We propose that immunotherapy is a plausible and practical therapeutic strategy and advocate more studies in this area to develop effective measures to control and treat these devastating disorders.
Collapse
Affiliation(s)
| | - Jiyan Ma
- Center for Neurodegenerative Science, Van Andel Institute, 333 Bostwick Avenue N.E., Grand Rapids, MI 49503, USA;
| |
Collapse
|
31
|
Frontzek K, Carta M, Losa M, Epskamp M, Meisl G, Anane A, Brandel JP, Camenisch U, Castilla J, Haïk S, Knowles T, Lindner E, Lutterotti A, Minikel EV, Roiter I, Safar JG, Sanchez-Valle R, Žáková D, Hornemann S, Aguzzi A. Autoantibodies against the prion protein in individuals with PRNP mutations. Neurology 2020; 95:e2028-e2037. [PMID: 32098855 DOI: 10.1212/wnl.0000000000009183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To determine whether naturally occurring autoantibodies against the prion protein are present in individuals with genetic prion disease mutations and controls, and if so, whether they are protective against prion disease. METHODS In this case-control study, we collected 124 blood samples from individuals with a variety of pathogenic PRNP mutations and 78 control individuals with a positive family history of genetic prion disease but lacking disease-associated PRNP mutations. Antibody reactivity was measured using an indirect ELISA for the detection of human immunoglobulin G1-4 antibodies against wild-type human prion protein. Multivariate linear regression models were constructed to analyze differences in autoantibody reactivity between (1) PRNP mutation carriers vs controls and (2) asymptomatic vs symptomatic PRNP mutation carriers. Robustness of results was examined in matched cohorts. RESULTS We found that antibody reactivity was present in a subset of both PRNP mutation carriers and controls. Autoantibody levels were not influenced by PRNP mutation status or clinical manifestation of prion disease. Post hoc analyses showed anti-PrPC autoantibody titers to be independent of personal history of autoimmune disease and other immunologic disorders, as well as PRNP codon 129 polymorphism. CONCLUSIONS Pathogenic PRNP variants do not notably stimulate antibody-mediated anti-PrPC immunity. Anti-PrPC immunoglobulin G autoantibodies are not associated with the onset of prion disease. The presence of anti-PrPC autoantibodies in the general population without any disease-specific association suggests that relatively high titers of naturally occurring antibodies are well-tolerated. CLINICALTRIALSGOV IDENTIFIER NCT02837705.
Collapse
Affiliation(s)
- Karl Frontzek
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia.
| | - Manfredi Carta
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Marco Losa
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Mirka Epskamp
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Georg Meisl
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Alice Anane
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Jean-Philippe Brandel
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Ulrike Camenisch
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Joaquín Castilla
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Stéphane Haïk
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Tuomas Knowles
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Ewald Lindner
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Andreas Lutterotti
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Eric Vallabh Minikel
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Ignazio Roiter
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Jiri G Safar
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Raquel Sanchez-Valle
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Dana Žáková
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Simone Hornemann
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Adriano Aguzzi
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia.
| | | |
Collapse
|
32
|
Krance SH, Luke R, Shenouda M, Israwi AR, Colpitts SJ, Darwish L, Strauss M, Watts JC. Cellular models for discovering prion disease therapeutics: Progress and challenges. J Neurochem 2020; 153:150-172. [PMID: 31943194 DOI: 10.1111/jnc.14956] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Abstract
Prions, which cause fatal neurodegenerative disorders such as Creutzfeldt-Jakob disease, are misfolded and infectious protein aggregates. Currently, there are no treatments available to halt or even delay the progression of prion disease in the brain. The infectious nature of prions has resulted in animal paradigms that accurately recapitulate all aspects of prion disease, and these have proven to be instrumental for testing the efficacy of candidate therapeutics. Nonetheless, infection of cultured cells with prions provides a much more powerful system for identifying molecules capable of interfering with prion propagation. Certain lines of cultured cells can be chronically infected with various types of mouse prions, and these models have been used to unearth candidate anti-prion drugs that are at least partially efficacious when administered to prion-infected rodents. However, these studies have also revealed that not all types of prions are equal, and that drugs active against mouse prions are not necessarily effective against prions from other species. Despite some recent progress, the number of cellular models available for studying non-mouse prions remains limited. In particular, human prions have proven to be particularly challenging to propagate in cultured cells, which has severely hindered the discovery of drugs for Creutzfeldt-Jakob disease. In this review, we summarize the cellular models that are presently available for discovering and testing drugs capable of blocking the propagation of prions and highlight challenges that remain on the path towards developing therapies for prion disease.
Collapse
Affiliation(s)
- Saffire H Krance
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Russell Luke
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Marc Shenouda
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Ahmad R Israwi
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Sarah J Colpitts
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Lina Darwish
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Maximilian Strauss
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
Holec SA, Block AJ, Bartz JC. The role of prion strain diversity in the development of successful therapeutic treatments. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:77-119. [PMID: 32958242 PMCID: PMC8939712 DOI: 10.1016/bs.pmbts.2020.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Prions are a self-propagating misfolded conformation of a cellular protein. Prions are found in several eukaryotic organisms with mammalian prion diseases encompassing a wide range of disorders. The first recognized prion disease, the transmissible spongiform encephalopathies (TSEs), affect several species including humans. Alzheimer's disease, synucleinopathies, and tauopathies share a similar mechanism of self-propagation of the prion form of the disease-specific protein reminiscent of the infection process of TSEs. Strain diversity in prion disease is characterized by differences in the phenotype of disease that is hypothesized to be encoded by strain-specific conformations of the prion form of the disease-specific protein. Prion therapeutics that target the prion form of the disease-specific protein can lead to the emergence of drug-resistant strains of prions, consistent with the hypothesis that prion strains exist as a dynamic mixture of a dominant strain in combination with minor substrains. To overcome this obstacle, therapies that reduce or eliminate the template of conversion are efficacious, may reverse neuropathology, and do not result in the emergence of drug resistance. Recent advancements in preclinical diagnosis of prion infection may allow for a combinational approach that treats the prion form and the precursor protein to effectively treat prion diseases.
Collapse
Affiliation(s)
- Sara A.M. Holec
- Institute for Applied Life Sciences and Department of Biology, University of Massachusetts Amherst, Amherst, MA, United States,Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE, United States
| | - Alyssa J. Block
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE, United States
| | - Jason C. Bartz
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE, United States,Corresponding author:
| |
Collapse
|
34
|
Abstract
Prion diseases are progressive, incurable and fatal neurodegenerative conditions. The term 'prion' was first nominated to express the revolutionary concept that a protein could be infectious. We now know that prions consist of PrPSc, the pathological aggregated form of the cellular prion protein PrPC. Over the years, the term has been semantically broadened to describe aggregates irrespective of their infectivity, and the prion concept is now being applied, perhaps overenthusiastically, to all neurodegenerative diseases that involve protein aggregation. Indeed, recent studies suggest that prion diseases (PrDs) and protein misfolding disorders (PMDs) share some common disease mechanisms, which could have implications for potential treatments. Nevertheless, the transmissibility of bona fide prions is unique, and PrDs should be considered as distinct from other PMDs.
Collapse
Affiliation(s)
- Claudia Scheckel
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
35
|
Cox TO, Gunther EC, Brody AH, Chiasseu MT, Stoner A, Smith LM, Haas LT, Hammersley J, Rees G, Dosanjh B, Groves M, Gardener M, Dobson C, Vaughan T, Chessell I, Billinton A, Strittmatter SM. Anti-PrP C antibody rescues cognition and synapses in transgenic alzheimer mice. Ann Clin Transl Neurol 2019; 6:554-574. [PMID: 30911579 PMCID: PMC6414488 DOI: 10.1002/acn3.730] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/28/2022] Open
Abstract
Objective Amyloid-beta oligomers (Aßo) trigger the development of Alzheimer's disease (AD) pathophysiology. Cellular prion protein (PrPC) initiates synaptic damage as a high affinity receptor for Aßo. Here, we evaluated the preclinical therapeutic efficacy of a fully human monoclonal antibody against PrPC. This AZ59 antibody selectively targets the Aβo binding site in the amino-terminal unstructured domain of PrPC to avoid any potential risk of direct toxicity. Methods Potency of AZ59 was evaluated by binding to PrPC, blockade of Aβo interaction and interruption of Aβo signaling. AZ59 was administered to mice by weekly intraperitoneal dosing and brain antibody measured. APP/PS1 transgenic mice were treated with AZ59 and assessed by memory tests, by brain biochemistry and by histochemistry for Aß, gliosis and synaptic density. Results AZ59 binds PrPC with 100 pmol/L affinity and blocks human brain Aßo binding to PrPC, as well as prevents synaptotoxic signaling. Weekly i.p. dosing of 20 mg/kg AZ59 in a murine form achieves trough brain antibody levels greater than 10 nmol/L. Aged symptomatic APP/PS1 transgenic mice treated with AZ59 for 5-7 weeks show a full rescue of behavioral and synaptic loss phenotypes. This recovery occurs without clearance of plaque pathology or elimination of gliosis. AZ59 treatment also normalizes synaptic signaling abnormalities in transgenic brain. These benefits are dose-dependent and persist for at least 1 month after the last dose. Interpretation Preclinical data demonstrate that systemic AZ59 therapy rescues central synapses and memory function from transgenic Alzheimer's disease pathology, supporting a disease-modifying therapeutic potential.
Collapse
Affiliation(s)
- Timothy O. Cox
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Erik C. Gunther
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - A. Harrison Brody
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Marius T. Chiasseu
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Austin Stoner
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Levi M. Smith
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Laura T. Haas
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Jayne Hammersley
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Gareth Rees
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Bhupinder Dosanjh
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Maria Groves
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Matthew Gardener
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Claire Dobson
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Tristan Vaughan
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Iain Chessell
- NeuroscienceIMED Biotech UnitAstraZenecaGranta ParkCambridgeCB21 6GHUK
| | - Andrew Billinton
- NeuroscienceIMED Biotech UnitAstraZenecaGranta ParkCambridgeCB21 6GHUK
| | - Stephen M. Strittmatter
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| |
Collapse
|
36
|
Abstract
Prion diseases are rapidly progressive, incurable neurodegenerative disorders caused by misfolded, aggregated proteins known as prions, which are uniquely infectious. Remarkably, these infectious proteins have been responsible for widespread disease epidemics, including kuru in humans, bovine spongiform encephalopathy in cattle, and chronic wasting disease in cervids, the latter of which has spread across North America and recently appeared in Norway and Finland. The hallmark histopathological features include widespread spongiform encephalopathy, neuronal loss, gliosis, and deposits of variably sized aggregated prion protein, ranging from small, soluble oligomers to long, thin, unbranched fibrils, depending on the disease. Here, we explore recent advances in prion disease research, from the function of the cellular prion protein to the dysfunction triggering neurotoxicity, as well as mechanisms underlying prion spread between cells. We also highlight key findings that have revealed new therapeutic targets and consider unanswered questions for future research.
Collapse
Affiliation(s)
- Christina J Sigurdson
- Departments of Pathology and Medicine, UC San Diego School of Medicine, University of California, San Diego, La Jolla, California 92093, USA;
| | - Jason C Bartz
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, Nebraska 68178, USA
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
37
|
Le NTT, Wu B, Harris DA. Prion neurotoxicity. Brain Pathol 2019; 29:263-277. [PMID: 30588688 DOI: 10.1111/bpa.12694] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/28/2018] [Indexed: 01/04/2023] Open
Abstract
Although the mechanisms underlying prion propagation and infectivity are now well established, the processes accounting for prion toxicity and pathogenesis have remained mysterious. These processes are of enormous clinical relevance as they hold the key to identification of new molecular targets for therapeutic intervention. In this review, we will discuss two broad areas of investigation relevant to understanding prion neurotoxicity. The first is the use of in vitro experimental systems that model key events in prion pathogenesis. In this context, we will describe a hippocampal neuronal culture system we developed that reproduces the earliest pathological alterations in synaptic morphology and function in response to PrPSc . This system has allowed us to define a core synaptotoxic signaling pathway involving the activation of NMDA and AMPA receptors, stimulation of p38 MAPK phosphorylation and collapse of the actin cytoskeleton in dendritic spines. The second area concerns a striking and unexpected phenomenon in which certain structural manipulations of the PrPC molecule itself, including introduction of N-terminal deletion mutations or binding of antibodies to C-terminal epitopes, unleash powerful toxic effects in cultured cells and transgenic mice. We will describe our studies of this phenomenon, which led to the recognition that it is related to the induction of large, abnormal ionic currents by the structurally altered PrP molecules. Our results suggest a model in which the flexible N-terminal domain of PrPC serves as a toxic effector which is regulated by intramolecular interactions with the globular C-terminal domain. Taken together, these two areas of study have provided important clues to underlying cellular and molecular mechanisms of prion neurotoxicity. Nevertheless, much remains to be done on this next frontier of prion science.
Collapse
Affiliation(s)
- Nhat T T Le
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Bei Wu
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - David A Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| |
Collapse
|
38
|
A bispecific immunotweezer prevents soluble PrP oligomers and abolishes prion toxicity. PLoS Pathog 2018; 14:e1007335. [PMID: 30273408 PMCID: PMC6181439 DOI: 10.1371/journal.ppat.1007335] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 10/11/2018] [Accepted: 09/13/2018] [Indexed: 11/28/2022] Open
Abstract
Antibodies to the prion protein, PrP, represent a promising therapeutic approach against prion diseases but the neurotoxicity of certain anti-PrP antibodies has caused concern. Here we describe scPOM-bi, a bispecific antibody designed to function as a molecular prion tweezer. scPOM-bi combines the complementarity-determining regions of the neurotoxic antibody POM1 and the neuroprotective POM2, which bind the globular domain (GD) and flexible tail (FT) respectively. We found that scPOM-bi confers protection to prion-infected organotypic cerebellar slices even when prion pathology is already conspicuous. Moreover, scPOM-bi prevents the formation of soluble oligomers that correlate with neurotoxic PrP species. Simultaneous targeting of both GD and FT was more effective than concomitant treatment with the individual molecules or targeting the tail alone, possibly by preventing the GD from entering a toxic-prone state. We conclude that simultaneous binding of the GD and flexible tail of PrP results in strong protection from prion neurotoxicity and may represent a promising strategy for anti-prion immunotherapy. Antibody immunotherapy is considered a viable strategy against prion disease. We previously showed that antibodies against the so-called globular domain of Prion Protein (PrP) can cause PrP dependent neurotoxicity; this does not happen for antibodies against the flexible tail of PrP, which therefore ought to be preferred for therapy. Here we show that simultaneous targeting of both globular domain and flexible tail by a bispecific, combination of a toxic and a non-toxic antibody, results in stronger protection against prion toxicity, even if the bispecific is administered when prion pathology is already conspicuous. We hypothesize that neurotoxicity arises from binding to specific “toxicity triggering sites” in the globular domain. We designed our bispecific with two aims: i) occupying one such site and preventing prion or other factors from docking to it and ii) binding to the flexible tail to engage the region of PrP necessary for neurotoxicity. We also show that neurotoxic antibodies cause the formation of soluble PrP oligomers that cause toxicity on PrP expressing cell lines; these are not formed in the presence of prion protective antibodies. We suggest that these soluble species might play a role in prion toxicity, similarly to what is generally agreed to happen in other neurodegenerative disorders.
Collapse
|
39
|
A Bioluminescent Cell Assay to Quantify Prion Protein Dimerization. Sci Rep 2018; 8:14178. [PMID: 30242186 PMCID: PMC6155003 DOI: 10.1038/s41598-018-32581-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 09/12/2018] [Indexed: 11/19/2022] Open
Abstract
The prion protein (PrP) is a cell surface protein that in disease misfolds and becomes infectious causing Creutzfeldt-Jakob disease in humans, scrapie in sheep, and chronic wasting disease in deer and elk. Little is known regarding the dimerization of PrP and its role in disease. We developed a bioluminescent prion assay (BPA) to quantify PrP dimerization by bimolecular complementation of split Gaussia luciferase (GLuc) halves that are each fused to PrP. Fusion constructs between PrP and N- and C-terminal GLuc halves were expressed on the surface of RK13 cells (RK13-DC cells) and dimerized to yield a bioluminescent signal that was decreased in the presence of eight different antibodies to PrP. Dimerization of PrP was independent of divalent cations and was induced under stress. Challenge of RK13-DC cells with seven different prion strains did not lead to detectable infection but was measurable by bioluminescence. Finally, we used BPA to screen a compound library for compounds inhibiting PrP dimerization. One of the most potent compounds to inhibit PrP dimerization was JTC-801, which also inhibited prion replication in RML-infected ScN2a and SMB cells with an EC50 of 370 nM and 220 nM, respectively. We show here that BPA is a versatile tool to study prion biology and to identify anti-prion compounds.
Collapse
|
40
|
Fang C, Wu B, Le NTT, Imberdis T, Mercer RCC, Harris DA. Prions activate a p38 MAPK synaptotoxic signaling pathway. PLoS Pathog 2018; 14:e1007283. [PMID: 30235355 PMCID: PMC6147624 DOI: 10.1371/journal.ppat.1007283] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 08/15/2018] [Indexed: 11/19/2022] Open
Abstract
Synaptic degeneration is one of the earliest pathological correlates of prion disease, and it is a major determinant of the progression of clinical symptoms. However, the cellular and molecular mechanisms underlying prion synaptotoxicity are poorly understood. Previously, we described an experimental system in which treatment of cultured hippocampal neurons with purified PrPSc, the infectious form of the prion protein, induces rapid retraction of dendritic spines, an effect that is entirely dependent on expression of endogenous PrPC by the target neurons. Here, we use this system to dissect pharmacologically the underlying cellular and molecular mechanisms. We show that PrPSc initiates a stepwise synaptotoxic signaling cascade that includes activation of NMDA receptors, calcium influx, stimulation of p38 MAPK and several downstream kinases, and collapse of the actin cytoskeleton within dendritic spines. Synaptic degeneration is restricted to excitatory synapses, spares presynaptic structures, and results in decrements in functional synaptic transmission. Pharmacological inhibition of any one of the steps in the signaling cascade, as well as expression of a dominant-negative form of p38 MAPK, block PrPSc-induced spine degeneration. Moreover, p38 MAPK inhibitors actually reverse the degenerative process after it has already begun. We also show that, while PrPC mediates the synaptotoxic effects of both PrPSc and the Alzheimer’s Aβ peptide in this system, the two species activate distinct signaling pathways. Taken together, our results provide powerful insights into the biology of prion neurotoxicity, they identify new, druggable therapeutic targets, and they allow comparison of prion synaptotoxic pathways with those involved in other neurodegenerative diseases. Prion diseases are a group of fatal neurodegenerative disorders that includes Creutzfeldt-Jakob disease and kuru in humans, and bovine spongiform encephalopathy in cattle. The infectious agent, or prion, that transmits these diseases is a naked protein molecule, the prion protein (PrP), which is an altered form of a normal, cellular protein. Although a great deal is known about how prions propagate themselves and transmit infection, the process by which they actually cause neurons to degenerate has remained mysterious. Here, we have used a specialized neuronal culture system to dissect the cellular and molecular mechanisms by which prions damage synapses, the structures that connect nerve cells and that play a crucial role in learning, memory, and neurological disease. Our results define a stepwise molecular pathway underlying prion synaptic toxicity, which involves activation of glutamate neurotransmitter receptors, influx of calcium ions into the neuron, and stimulation of specific mitogen-activated protein kinases, which attach phosphate groups to proteins to regulate their activity. We demonstrate that specific drugs, as well as a dominant-negative kinase mutant, block these steps and thereby prevent the synaptic degeneration produced by prions. Our results provide new insights into the pathogenesis of prion diseases, they uncover new drug targets for treating these diseases, and they allow us to compare prion diseases to other, more common neurodegenerative disorders like Alzheimer’s disease.
Collapse
Affiliation(s)
- Cheng Fang
- Department of Biochemistry, Boston University School of Medicine, Boston MA, United States of America
| | - Bei Wu
- Department of Biochemistry, Boston University School of Medicine, Boston MA, United States of America
| | - Nhat T. T. Le
- Department of Biochemistry, Boston University School of Medicine, Boston MA, United States of America
| | - Thibaut Imberdis
- Department of Biochemistry, Boston University School of Medicine, Boston MA, United States of America
| | - Robert C. C. Mercer
- Department of Biochemistry, Boston University School of Medicine, Boston MA, United States of America
| | - David A. Harris
- Department of Biochemistry, Boston University School of Medicine, Boston MA, United States of America
- * E-mail:
| |
Collapse
|
41
|
Hirsch TZ, Martin-Lannerée S, Reine F, Hernandez-Rapp J, Herzog L, Dron M, Privat N, Passet B, Halliez S, Villa-Diaz A, Lacroux C, Klein V, Haïk S, Andréoletti O, Torres JM, Vilotte JL, Béringue V, Mouillet-Richard S. Epigenetic Control of the Notch and Eph Signaling Pathways by the Prion Protein: Implications for Prion Diseases. Mol Neurobiol 2018; 56:2159-2173. [PMID: 29998397 DOI: 10.1007/s12035-018-1193-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 06/26/2018] [Indexed: 12/12/2022]
Abstract
Among the ever-growing number of self-replicating proteins involved in neurodegenerative diseases, the prion protein PrP remains the most infamous for its central role in transmissible spongiform encephalopathies (TSEs). In these diseases, pathogenic prions propagate through a seeding mechanism, where normal PrPC molecules are converted into abnormally folded scrapie isoforms termed PrPSc. Since its discovery over 30 years ago, much advance has contributed to define the host-encoded cellular prion protein PrPC as a critical relay of prion-induced neuronal cell demise. A current consensual view is that the conversion of PrPC into PrPSc in neuronal cells diverts the former from its normal function with subsequent molecular alterations affecting synaptic plasticity. Here, we report that prion infection is associated with reduced expression of key effectors of the Notch pathway in vitro and in vivo, recapitulating changes fostered by the absence of PrPC. We further show that both prion infection and PrPC depletion promote drastic alterations in the expression of a defined set of Eph receptors and their ephrin ligands, which represent important players in synaptic function. Our data indicate that defects in the Notch and Eph axes can be mitigated in response to histone deacetylase inhibition in PrPC-depleted as well as prion-infected cells. We thus conclude that infectious prions cause a loss-of-function phenotype with respect to Notch and Eph signaling and that these alterations are sustained by epigenetic mechanisms.
Collapse
Affiliation(s)
- Théo Z Hirsch
- INSERM UMR 1124, 75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, 75006, Paris, France
- INSERM U1162, 75010, Paris, France
| | - Séverine Martin-Lannerée
- INSERM UMR 1124, 75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, 75006, Paris, France
| | - Fabienne Reine
- INRA, Université Paris-Saclay, UR 892 Virologie Immunologie Moléculaires, 78350, Jouy-en-Josas, France
| | - Julia Hernandez-Rapp
- INSERM UMR 1124, 75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, 75006, Paris, France
- Centre de Recherche du CHU de Québec, Université Laval, Québec, G1V4G2, Québec, Canada
| | - Laetitia Herzog
- INRA, Université Paris-Saclay, UR 892 Virologie Immunologie Moléculaires, 78350, Jouy-en-Josas, France
| | - Michel Dron
- INRA, Université Paris-Saclay, UR 892 Virologie Immunologie Moléculaires, 78350, Jouy-en-Josas, France
| | - Nicolas Privat
- INSERM UMR 1127, CNRS UMR 7225, 75013, Paris, France
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013, Paris, France
| | - Bruno Passet
- INRA UMR1313, Génétique Animale et Biologie Intégrative, 78350, Jouy-en-Josas, France
| | - Sophie Halliez
- INRA, Université Paris-Saclay, UR 892 Virologie Immunologie Moléculaires, 78350, Jouy-en-Josas, France
- INSERM, UMR-S1172, Lille University, 59045, Lille, France
| | - Ana Villa-Diaz
- Centro de Investigación en Sanidad Animal-INIA, 28130, Madrid, Spain
| | | | - Victor Klein
- INSERM UMR 1124, 75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, 75006, Paris, France
| | - Stéphane Haïk
- INSERM UMR 1127, CNRS UMR 7225, 75013, Paris, France
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013, Paris, France
| | | | - Juan-Maria Torres
- Centro de Investigación en Sanidad Animal-INIA, 28130, Madrid, Spain
| | - Jean-Luc Vilotte
- INRA UMR1313, Génétique Animale et Biologie Intégrative, 78350, Jouy-en-Josas, France
| | | | - Sophie Mouillet-Richard
- INSERM UMR 1124, 75006, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, 75006, Paris, France.
| |
Collapse
|
42
|
Baral PK, Swayampakula M, Aguzzi A, James MNG. Structural characterization of
POM
6 Fab and mouse prion protein complex identifies key regions for prions conformational conversion. FEBS J 2018; 285:1701-1714. [DOI: 10.1111/febs.14438] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 02/02/2018] [Accepted: 03/16/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Pravas Kumar Baral
- Department of Biochemistry Faculty of Medicine and Dentistry University of Alberta Edmonton Canada
| | - Mridula Swayampakula
- Department of Biochemistry Faculty of Medicine and Dentistry University of Alberta Edmonton Canada
| | - Adriano Aguzzi
- Institute of Neuropathology University of Zurich Switzerland
| | - Michael N. G. James
- Department of Biochemistry Faculty of Medicine and Dentistry University of Alberta Edmonton Canada
| |
Collapse
|
43
|
Leighton PLA, Allison WT. Protein Misfolding in Prion and Prion-Like Diseases: Reconsidering a Required Role for Protein Loss-of-Function. J Alzheimers Dis 2018; 54:3-29. [PMID: 27392869 DOI: 10.3233/jad-160361] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Prion disease research has contributed much toward understanding other neurodegenerative diseases, including recent demonstrations that Alzheimer's disease (AD) and other neurodegenerative diseases are prion-like. Prion-like diseases involve the spread of degeneration between individuals and/or among cells or tissues via template directed misfolding, wherein misfolded protein conformers propagate disease by causing normal proteins to misfold. Here we use the premise that AD, amyotrophic lateral sclerosis, Huntington's disease, and other similar diseases are prion-like and ask: Can we apply knowledge gained from studies of these prion-like diseases to resolve debates about classical prion diseases? We focus on controversies about what role(s) protein loss-of-function might have in prion diseases because this has therapeutic implications, including for AD. We examine which loss-of-function events are recognizable in prion-like diseases by considering the normal functions of the proteins before their misfolding and aggregation. We then delineate scenarios wherein gain-of-function and/or loss-of-function would be necessary or sufficient for neurodegeneration. We consider roles of PrPC loss-of-function in prion diseases and in AD, and conclude that the conventional wisdom that prion diseases are 'toxic gain-of-function diseases' has limitations. While prion diseases certainly have required gain-of-function components, we propose that disease phenotypes are predominantly caused by deficits in the normal physiology of PrPC and its interaction partners as PrPC converts to PrPSc. In this model, gain-of-function serves mainly to spread disease, and loss-of-function directly mediates neuron dysfunction. We propose experiments and predictions to assess our conclusion. Further study on the normal physiological roles of these key proteins is warranted.
Collapse
Affiliation(s)
- Patricia L A Leighton
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - W Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
44
|
Taschuk R, Scruten E, Woodbury M, Cashman N, Potter A, Griebel P, Tikoo SK, Napper S. Induction of PrP Sc-specific systemic and mucosal immune responses in white-tailed deer with an oral vaccine for chronic wasting disease. Prion 2018; 11:368-380. [PMID: 28968152 PMCID: PMC5639826 DOI: 10.1080/19336896.2017.1367083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The ongoing epidemic of chronic wasting disease (CWD) within cervid populations indicates the need for novel approaches for disease management. A vaccine that either reduces susceptibility to infection or reduces shedding of prions by infected animals, or a combination of both, could be of benefit for disease control. The development of such a vaccine is challenged by the unique nature of prion diseases and the requirement for formulation and delivery in an oral format for application in wildlife settings. To address the unique nature of prions, our group targets epitopes, termed disease specific epitopes (DSEs), whose exposure for antibody binding depends on disease-associated misfolding of PrPC into PrPSc. Here, a DSE corresponding to the rigid loop (RL) region, which was immunogenic following parenteral vaccination, was translated into an oral vaccine. This vaccine consists of a replication-incompetent human adenovirus expressing a truncated rabies glycoprotein G recombinant fusion with the RL epitope (hAd5:tgG-RL). Oral immunization of white-tailed deer with hAd5:tgG-RL induced PrPSc-specific systemic and mucosal antibody responses with an encouraging safety profile in terms of no adverse health effects nor prolonged vector shedding. By building upon proven strategies of formulation for wildlife vaccines, these efforts generate a particular PrPSc-specific oral vaccine for CWD as well as providing a versatile platform, in terms of carrier protein and biological vector, for generation of other oral, peptide-based CWD vaccines.
Collapse
Affiliation(s)
- Ryan Taschuk
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada.,b School of Public Health, University of Saskatchewan , Saskatoon, Saskatchewan , Canada
| | - Erin Scruten
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada
| | - Murray Woodbury
- c Western College of Veterinary Medicine, University of Saskatchewan , Saskatoon , Saskatchewan , Canada
| | - Neil Cashman
- d Department of Neurology , University of British Columbia , Vancouver , BC , Canada
| | - Andrew Potter
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada
| | - Philip Griebel
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada.,b School of Public Health, University of Saskatchewan , Saskatoon, Saskatchewan , Canada
| | - Suresh K Tikoo
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada.,b School of Public Health, University of Saskatchewan , Saskatoon, Saskatchewan , Canada
| | - Scott Napper
- a Vaccine and Infectious Disease Organization, University of Saskatchewan , Saskatoon , Saskatchewan , Canada.,e Department of Biochemistry , University of Saskatchewan , Saskatoon , Saskatchewan , Canada
| |
Collapse
|
45
|
McDonald AJ, Wu B, Harris DA. An inter-domain regulatory mechanism controls toxic activities of PrP C. Prion 2017; 11:388-397. [PMID: 28960140 DOI: 10.1080/19336896.2017.1384894] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The normal function of PrPC, the cellular prion protein, has remained mysterious since its first description over 30 years ago. Amazingly, although complete deletion of the gene encoding PrPC has little phenotypic consequence, expression in transgenic mice of PrP molecules carrying certain internal deletions produces dramatic neurodegenerative phenotypes. In our recent paper, 1 we have demonstrated that the flexible, N-terminal domain of PrPC possesses toxic effector functions, which are regulated by a docking interaction with the structured, C-terminal domain. Disruption of this inter-domain interaction, for example by deletions of the hinge region or by binding of antibodies to the C-terminal domain, results in abnormal ionic currents and degeneration of dendritic spines in cultured neuronal cells. This mechanism may contribute to the neurotoxicity of PrPSc and possibly other protein aggregates, and could play a role in the physiological activity of PrPC. These results also provide a warning about the potential toxic side effects of PrP-directed antibody therapies for prion and Alzheimer's diseases.
Collapse
Affiliation(s)
- Alex J McDonald
- a Department of Biochemistry , Boston University School of Medicine , Boston , MA , USA
| | - Bei Wu
- a Department of Biochemistry , Boston University School of Medicine , Boston , MA , USA
| | - David A Harris
- a Department of Biochemistry , Boston University School of Medicine , Boston , MA , USA
| |
Collapse
|
46
|
Sorce S, Stocker R, Seredenina T, Holmdahl R, Aguzzi A, Chio A, Depaulis A, Heitz F, Olofsson P, Olsson T, Duveau V, Sanoudou D, Skosgater S, Vlahou A, Wasquel D, Krause KH, Jaquet V. NADPH oxidases as drug targets and biomarkers in neurodegenerative diseases: What is the evidence? Free Radic Biol Med 2017; 112:387-396. [PMID: 28811143 DOI: 10.1016/j.freeradbiomed.2017.08.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 08/04/2017] [Accepted: 08/06/2017] [Indexed: 11/25/2022]
Abstract
Neurodegenerative disease are frequently characterized by microglia activation and/or leukocyte infiltration in the parenchyma of the central nervous system and at the molecular level by increased oxidative modifications of proteins, lipids and nucleic acids. NADPH oxidases (NOX) emerged as a novel promising class of pharmacological targets for the treatment of neurodegeneration due to their role in oxidant generation and presumably in regulating microglia activation. The unique function of NOX is the generation of superoxide anion (O2•-) and hydrogen peroxide (H2O2). However in the context of neuroinflammation, they present paradoxical features since O2•-/H2O2 generated by NOX and/or secondary reactive oxygen species (ROS) derived from O2•-/H2O2 can either lead to neuronal oxidative damage or resolution of inflammation. The role of NOX enzymes has been investigated in many models of neurodegenerative diseases by using either genetic or pharmacological approaches. In the present review we provide a critical assessment of recent findings related to the role of NOX in the CNS as well as how the field has advanced over the last 5 years. In particular, we focus on the data derived from the work of a consortium (Neurinox) funded by the European Commission's Programme 7 (FP7). We discuss the evidence gathered from animal models and human samples linking NOX expression/activity with neuroinflammation in neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS) and Creutzfeldt-Jakob disease as well as autoimmune demyelinating diseases like multiple sclerosis (MS) and chronic inflammatory demyelinating polyneuropathy (CIDP). We address the possibility to use measurement of the activity of the NOX2 isoform in blood samples as biomarker of disease severity and treatment efficacy in neurodegenerative disease. Finally we clarify key controversial aspects in the field of NOX, such as NOX cellular expression in the brain, measurement of NOX activity, impact of genetic deletion of NOX in animal models of neurodegeneration and specificity of NOX inhibitors.
Collapse
Affiliation(s)
- Silvia Sorce
- Neuropathology Institute, University of Zürich, Switzerland
| | - Roland Stocker
- Victor Chang Cardiac Research Institute, and School of Medical Sciences, University of New South Wales, Australia
| | - Tamara Seredenina
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - Rikard Holmdahl
- Section for Medical Inflammation research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Sweden
| | - Adriano Aguzzi
- Neuropathology Institute, University of Zürich, Switzerland
| | - Adriano Chio
- Department of Neuroscience, University of Torino, Italy
| | - Antoine Depaulis
- Grenoble Institut des Neurosciences, Inserm U1216 and Univ, Grenoble Alpes, F-38000 Grenoble, France
| | | | - Peter Olofsson
- Redoxis AB, Medicon Village, Lund, Sweden; Pronoxis AB, Medicon Village, Lund, Sweden
| | - Tomas Olsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Sweden
| | | | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sara Skosgater
- Arttic, 58A rue du Dessous des Berges, F-75013 Paris, France
| | - Antonia Vlahou
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | - Karl-Heinz Krause
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - Vincent Jaquet
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland.
| |
Collapse
|
47
|
Goniotaki D, Lakkaraju AKK, Shrivastava AN, Bakirci P, Sorce S, Senatore A, Marpakwar R, Hornemann S, Gasparini F, Triller A, Aguzzi A. Inhibition of group-I metabotropic glutamate receptors protects against prion toxicity. PLoS Pathog 2017; 13:e1006733. [PMID: 29176838 PMCID: PMC5720820 DOI: 10.1371/journal.ppat.1006733] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/07/2017] [Accepted: 11/04/2017] [Indexed: 12/29/2022] Open
Abstract
Prion infections cause inexorable, progressive neurological dysfunction and neurodegeneration. Expression of the cellular prion protein PrPC is required for toxicity, suggesting the existence of deleterious PrPC-dependent signaling cascades. Because group-I metabotropic glutamate receptors (mGluR1 and mGluR5) can form complexes with the cellular prion protein (PrPC), we investigated the impact of mGluR1 and mGluR5 inhibition on prion toxicity ex vivo and in vivo. We found that pharmacological inhibition of mGluR1 and mGluR5 antagonized dose-dependently the neurotoxicity triggered by prion infection and by prion-mimetic anti-PrPC antibodies in organotypic brain slices. Prion-mimetic antibodies increased mGluR5 clustering around dendritic spines, mimicking the toxicity of Aβ oligomers. Oral treatment with the mGluR5 inhibitor, MPEP, delayed the onset of motor deficits and moderately prolonged survival of prion-infected mice. Although group-I mGluR inhibition was not curative, these results suggest that it may alleviate the neurological dysfunctions induced by prion diseases.
Collapse
Affiliation(s)
| | | | - Amulya N. Shrivastava
- École Normale Supérieure, Institut de Biologie de l'ENS (IBENS) INSERM CNRS PSL Research University, Paris, France
- Paris-Saclay Institute of Neuroscience, CNRS, Gif-sur-Yvette, France
| | - Pamela Bakirci
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Silvia Sorce
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Assunta Senatore
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | | | - Simone Hornemann
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | | | - Antoine Triller
- École Normale Supérieure, Institut de Biologie de l'ENS (IBENS) INSERM CNRS PSL Research University, Paris, France
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
48
|
Abstract
Three decades after the discovery of prions as the cause of Creutzfeldt-Jakob disease and other transmissible spongiform encephalopathies, we are still nowhere close to finding an effective therapy. Numerous pharmacological interventions have attempted to target various stages of disease progression, yet none has significantly ameliorated the course of disease. We still lack a mechanistic understanding of how the prions damage the brain, and this situation results in a dearth of validated pharmacological targets. In this review, we discuss the attempts to interfere with the replication of prions and to enhance their clearance. We also trace some of the possibilities to identify novel targets that may arise with increasing insights into prion biology.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland;
| | - Asvin K K Lakkaraju
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland;
| | - Karl Frontzek
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland;
| |
Collapse
|
49
|
Copper- and Zinc-Promoted Interdomain Structure in the Prion Protein: A Mechanism for Autoinhibition of the Neurotoxic N-Terminus. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:35-56. [PMID: 28838668 DOI: 10.1016/bs.pmbts.2017.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The function of the cellular prion protein (PrPC), while still poorly understood, is increasingly linked to its ability to bind physiological metal ions at the cell surface. PrPC binds divalent forms of both copper and zinc through its unstructured N-terminal domain, modulating interactions between PrPC and various receptors at the cell surface and ultimately tuning downstream cellular processes. In this chapter, we briefly discuss the molecular features of copper and zinc uptake by PrPC and summarize evidence implicating these metal ions in PrP-mediated physiology. We then focus our review on recent biophysical evidence revealing a physical interaction between the flexible N-terminal and globular C-terminal domains of PrPC. This interdomain cis interaction is electrostatic in nature and is promoted by the binding of Cu2+ and Zn2+ to the N-terminal octarepeat domain. These findings, along with recent cellular studies, suggest a mechanism whereby NC interactions serve to regulate the activity and/or toxicity of the PrPC N-terminus. We discuss this potential mechanism in relation to familial prion disease mutations, lethal deletions of the PrPC central region, and neurotoxicity induced by certain globular domain ligands, including bona fide prions and toxic amyloid-β oligomers.
Collapse
|
50
|
Hirsch TZ, Martin-Lannerée S, Mouillet-Richard S. Functions of the Prion Protein. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:1-34. [PMID: 28838656 DOI: 10.1016/bs.pmbts.2017.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although initially disregarded compared to prion pathogenesis, the functions exerted by the cellular prion protein PrPC have gained much interest over the past two decades. Research aiming at unraveling PrPC functions started to intensify when it became appreciated that it would give clues as to how it is subverted in the context of prion infection and, more recently, in the context of Alzheimer's disease. It must now be admitted that PrPC is implicated in an incredible variety of biological processes, including neuronal homeostasis, stem cell fate, protection against stress, or cell adhesion. It appears that these diverse roles can all be fulfilled through the involvement of PrPC in cell signaling events. Our aim here is to provide an overview of our current understanding of PrPC functions from the animal to the molecular scale and to highlight some of the remaining gaps that should be addressed in future research.
Collapse
Affiliation(s)
- Théo Z Hirsch
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Séverine Martin-Lannerée
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Sophie Mouillet-Richard
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France.
| |
Collapse
|