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
|
Simmons SM, Payne VL, Hrdlicka JG, Taylor J, Larsen PA, Wolf TM, Schwabenlander MD, Yuan Q, Bartz JC. Rapid and sensitive determination of residual prion infectivity from prion-decontaminated surfaces. mSphere 2024; 9:e0050424. [PMID: 39189773 PMCID: PMC11423590 DOI: 10.1128/msphere.00504-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/21/2024] [Indexed: 08/28/2024] Open
Abstract
Prion diseases are untreatable fatal transmissible neurodegenerative diseases that affect a wide range of mammals, including humans, and are caused by PrPSc, the infectious self-templating conformation of the host-encoded protein, PrPC. Prion diseases can be transmitted via surfaces (e.g., forceps, EEG electrodes) in laboratory and clinical settings. Here, we use a combination of surface swabbing and real-time quaking-induced conversion (RT-QuIC) to test for residual surface-associated prions following prion disinfection. We found that treatment of several prion-contaminated laboratory and clinically relevant surfaces with either water or 70% EtOH resulted in robust detection of surface-associated prions. In contrast, treatment of surfaces with sodium hypochlorite resulted in a failure to detect surface-associated prions. RT-QuIC analysis of prion-contaminated stainless steel wires paralleled the findings of the surface swab studies. Importantly, animal bioassay and RT-QuIC analysis of the same swab extracts are in agreement. We report on conditions that may interfere with the assay that need to be taken into consideration before using this technique. Overall, this method can be used to survey laboratory and clinical surfaces for prion infectivity following prion decontamination protocols.IMPORTANCEPrion diseases can be accidentally transmitted in clinical and occupational settings. While effective means of prion decontamination exist, methods for determining the effectiveness are only beginning to be described. Here, we analyze surface swab extracts using real-time quaking-induced conversion (RT-QuIC) to test for residual prions following prion disinfection of relevant clinical and laboratory surfaces. We found that this method can rapidly determine the efficacy of surface prion decontamination. Importantly, examination of surface extracts with RT-QuIC and animal bioassay produced similar findings, suggesting that this method can accurately assess the reduction in prion titer. We identified surface contaminants that interfere with the assay, which may be found in clinical and laboratory settings. Overall, this method can enhance clinical and laboratory prion safety measures.
Collapse
Affiliation(s)
- Sara M. Simmons
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, Nebraska, USA
| | | | - Jay G. Hrdlicka
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, Nebraska, USA
| | - Jack Taylor
- Biostatistical Core Facility, Creighton University, Omaha, Nebraska, USA
| | - Peter A. Larsen
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
- Minnesota Center for Prion Research and Outreach, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Tiffany M. Wolf
- Minnesota Center for Prion Research and Outreach, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Marc D. Schwabenlander
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
- Minnesota Center for Prion Research and Outreach, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Qi Yuan
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, Nebraska, USA
| | - Jason C. Bartz
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, Nebraska, USA
- Minnesota Center for Prion Research and Outreach, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
- Prion Research Center, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
3
|
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
|
4
|
Thellung S, Corsaro A, Dellacasagrande I, Nizzari M, Zambito M, Florio T. Proteostasis unbalance in prion diseases: Mechanisms of neurodegeneration and therapeutic targets. Front Neurosci 2022; 16:966019. [PMID: 36148145 PMCID: PMC9485628 DOI: 10.3389/fnins.2022.966019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/05/2022] [Indexed: 01/18/2023] Open
Abstract
Transmissible spongiform encephalopathies (TSEs), or prion diseases, are progressive neurodegenerative disorders of the central nervous system that affect humans and animals as sporadic, inherited, and infectious forms. Similarly to Alzheimer's disease and other neurodegenerative disorders, any attempt to reduce TSEs' lethality or increase the life expectancy of affected individuals has been unsuccessful. Typically, the onset of symptoms anticipates the fatal outcome of less than 1 year, although it is believed to be the consequence of a decades-long process of neuronal death. The duration of the symptoms-free period represents by itself a major obstacle to carry out effective neuroprotective therapies. Prions, the infectious entities of TSEs, are composed of a protease-resistant protein named prion protein scrapie (PrPSc) from the prototypical TSE form that afflicts ovines. PrPSc misfolding from its physiological counterpart, cellular prion protein (PrPC), is the unifying pathogenic trait of all TSEs. PrPSc is resistant to intracellular turnover and undergoes amyloid-like fibrillation passing through the formation of soluble dimers and oligomers, which are likely the effective neurotoxic entities. The failure of PrPSc removal is a key pathogenic event that defines TSEs as proteopathies, likewise other neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's disease, characterized by alteration of proteostasis. Under physiological conditions, protein quality control, led by the ubiquitin-proteasome system, and macroautophagy clears cytoplasm from improperly folded, redundant, or aggregation-prone proteins. There is evidence that both of these crucial homeostatic pathways are impaired during the development of TSEs, although it is still unclear whether proteostasis alteration facilitates prion protein misfolding or, rather, PrPSc protease resistance hampers cytoplasmic protein quality control. This review is aimed to critically analyze the most recent advancements in the cause-effect correlation between PrPC misfolding and proteostasis alterations and to discuss the possibility that pharmacological restoring of ubiquitin-proteasomal competence and stimulation of autophagy could reduce the intracellular burden of PrPSc and ameliorate the severity of prion-associated neurodegeneration.
Collapse
Affiliation(s)
- Stefano Thellung
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Alessandro Corsaro
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Irene Dellacasagrande
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Mario Nizzari
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Martina Zambito
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Tullio Florio
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- *Correspondence: Tullio Florio
| |
Collapse
|
5
|
Dematteis G, Restelli E, Vanella VV, Manfredi M, Marengo E, Corazzari M, Genazzani AA, Chiesa R, Lim D, Tapella L. Calcineurin Controls Cellular Prion Protein Expression in Mouse Astrocytes. Cells 2022; 11:cells11040609. [PMID: 35203261 PMCID: PMC8870693 DOI: 10.3390/cells11040609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 02/05/2023] Open
Abstract
Prion diseases arise from the conformational conversion of the cellular prion protein (PrPC) into a self-replicating prion isoform (PrPSc). Although this process has been studied mostly in neurons, a growing body of evidence suggests that astrocytes express PrPC and are able to replicate and accumulate PrPSc. Currently, prion diseases remain incurable, while downregulation of PrPC represents the most promising therapy due to the reduction of the substrate for prion conversion. Here we show that the astrocyte-specific genetic ablation or pharmacological inhibition of the calcium-activated phosphatase calcineurin (CaN) reduces PrPC expression in astrocytes. Immunocytochemical analysis of cultured CaN-KO astrocytes and isolation of synaptosomal compartments from the hippocampi of astrocyte-specific CaN-KO (ACN-KO) mice suggest that PrPC is downregulated both in vitro and in vivo. The downregulation occurs without affecting the glycosylation of PrPC and without alteration of its proteasomal or lysosomal degradation. Direct assessment of the protein synthesis rate and shotgun mass spectrometry proteomics analysis suggest that the reduction of PrPC is related to the impairment of global protein synthesis in CaN-KO astrocytes. When WT-PrP and PrP-D177N, a mouse homologue of a human mutation associated with the inherited prion disease fatal familial insomnia, were expressed in astrocytes, CaN-KO astrocytes showed an aberrant localization of both WT-PrP and PrP-D177N variants with predominant localization to the Golgi apparatus, suggesting that ablation of CaN affects both WT and mutant PrP proteins. These results provide new mechanistic details in relation to the regulation of PrP expression in astrocytes, suggesting the therapeutic potential of astroglial cells.
Collapse
Affiliation(s)
- Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
| | - Elena Restelli
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (E.R.); (R.C.)
| | - Virginia Vita Vanella
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (V.V.V.); (M.M.)
| | - Marcello Manfredi
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (V.V.V.); (M.M.)
| | - Emilio Marengo
- Department of Sciences and Technological Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy;
| | - Marco Corazzari
- Department of Health Science (DSS), Center for Translational Research on Autoimmune and Allergic Disease (CAAD) & Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy;
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
| | - Roberto Chiesa
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (E.R.); (R.C.)
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
- Correspondence: (D.L.); (L.T.); Tel.: +39-0321-375822 (L.T.)
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
- Correspondence: (D.L.); (L.T.); Tel.: +39-0321-375822 (L.T.)
| |
Collapse
|
6
|
Chassefeyre R, Chaiamarit T, Verhelle A, Novak SW, Andrade LR, Leitão ADG, Manor U, Encalada SE. Endosomal sorting drives the formation of axonal prion protein endoggresomes. SCIENCE ADVANCES 2021; 7:eabg3693. [PMID: 34936461 PMCID: PMC8694590 DOI: 10.1126/sciadv.abg3693] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 11/05/2021] [Indexed: 05/15/2023]
Abstract
The pathogenic aggregation of misfolded prion protein (PrP) in axons underlies prion disease pathologies. The molecular mechanisms driving axonal misfolded PrP aggregate formation leading to neurotoxicity are unknown. We found that the small endolysosomal guanosine triphosphatase (GTPase) Arl8b recruits kinesin-1 and Vps41 (HOPS) onto endosomes carrying misfolded mutant PrP to promote their axonal entry and homotypic fusion toward aggregation inside enlarged endomembranes that we call endoggresomes. This axonal rapid endosomal sorting and transport-dependent aggregation (ARESTA) mechanism forms pathologic PrP endoggresomes that impair calcium dynamics and reduce neuronal viability. Inhibiting ARESTA diminishes endoggresome formation, rescues calcium influx, and prevents neuronal death. Our results identify ARESTA as a key pathway for the regulation of endoggresome formation and a new actionable antiaggregation target to ameliorate neuronal dysfunction in the prionopathies.
Collapse
Affiliation(s)
- Romain Chassefeyre
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tai Chaiamarit
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Adriaan Verhelle
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sammy Weiser Novak
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Leonardo R. Andrade
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - André D. G. Leitão
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Uri Manor
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sandra E. Encalada
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
7
|
Virus Infection, Genetic Mutations, and Prion Infection in Prion Protein Conversion. Int J Mol Sci 2021; 22:ijms222212439. [PMID: 34830321 PMCID: PMC8624980 DOI: 10.3390/ijms222212439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 01/04/2023] Open
Abstract
Conformational conversion of the cellular isoform of prion protein, PrPC, into the abnormally folded, amyloidogenic isoform, PrPSc, is an underlying pathogenic mechanism in prion diseases. The diseases manifest as sporadic, hereditary, and acquired disorders. Etiological mechanisms driving the conversion of PrPC into PrPSc are unknown in sporadic prion diseases, while prion infection and specific mutations in the PrP gene are known to cause the conversion of PrPC into PrPSc in acquired and hereditary prion diseases, respectively. We recently reported that a neurotropic strain of influenza A virus (IAV) induced the conversion of PrPC into PrPSc as well as formation of infectious prions in mouse neuroblastoma cells after infection, suggesting the causative role of the neuronal infection of IAV in sporadic prion diseases. Here, we discuss the conversion mechanism of PrPC into PrPSc in different types of prion diseases, by presenting our findings of the IAV infection-induced conversion of PrPC into PrPSc and by reviewing the so far reported transgenic animal models of hereditary prion diseases and the reverse genetic studies, which have revealed the structure-function relationship for PrPC to convert into PrPSc after prion infection.
Collapse
|
8
|
Lavigna G, Masone A, Bouybayoune I, Bertani I, Lucchetti J, Gobbi M, Porcu L, Zordan S, Rigamonti M, Imeri L, Restelli E, Chiesa R. Doxycycline rescues recognition memory and circadian motor rhythmicity but does not prevent terminal disease in fatal familial insomnia mice. Neurobiol Dis 2021; 158:105455. [PMID: 34358614 PMCID: PMC8463834 DOI: 10.1016/j.nbd.2021.105455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 01/15/2023] Open
Abstract
Fatal familial insomnia (FFI) is a dominantly inherited prion disease linked to the D178N mutation in the gene encoding the prion protein (PrP). Symptoms, including insomnia, memory loss and motor abnormalities, appear around 50 years of age, leading to death within two years. No treatment is available. A ten-year clinical trial of doxycycline (doxy) is under way in healthy individuals at risk of FFI to test whether presymptomatic doxy prevents or delays the onset of disease. To assess the drug's effect in a tractable disease model, we used Tg(FFI-26) mice, which accumulate aggregated and protease-resistant PrP in their brains and develop a fatal neurological illness highly reminiscent of FFI. Mice were treated daily with 10 mg/kg doxy starting from a presymptomatic stage for twenty weeks. Doxy rescued memory deficits and restored circadian motor rhythmicity in Tg(FFI-26) mice. However, it did not prevent the onset and progression of motor dysfunction, clinical signs and progression to terminal disease. Doxy did not change the amount of aggregated and protease-resistant PrP, but reduced microglial activation in the hippocampus. Presymptomatic doxy treatment rescues cognitive impairment and the motor correlates of sleep dysfunction in Tg(FFI-26) mice but does not prevent fatal disease.
Collapse
Affiliation(s)
- Giada Lavigna
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Antonio Masone
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ihssane Bouybayoune
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilaria Bertani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jacopo Lucchetti
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Gobbi
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luca Porcu
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | - Luca Imeri
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Elena Restelli
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Roberto Chiesa
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| |
Collapse
|
9
|
Shiraishi N, Hirano Y. Combination of Copper Ions and Nucleotide Generates Aggregates from Prion Protein Fragments in the N-Terminal Domain. Protein Pept Lett 2021; 27:782-792. [PMID: 32096738 DOI: 10.2174/0929866527666200225124829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND It has been previously found that PrP23-98, which contains four highly conserved octarepeats (residues 60-91) and one partial repeat (residues 92-96), polymerizes into amyloid-like and proteinase K-resistant spherical aggregates in the presence of NADPH plus copper ions. OBJECTIVE We aimed to determine the requirements for the formation of these aggregates. METHODS In this study, we performed an aggregation experiment using N-acetylated and Camidated PrP fragments of the N-terminal domain, Octa1, Octa2, Octa3, Octa4, PrP84-114, and PrP76-114, in the presence of NADPH with copper ions, and focused on the effect of the number of copper-binding sites on aggregation. RESULTS Among these PrP fragments, Octa4, containing four copper-binding sites, was particularly effective in forming aggregates. We also tested the effect of other pyridine nucleotides and adenine nucleotides on the aggregation of Octa4. ATP was equally effective, but NADH, NADP, ADP, and AMP had no effect. CONCLUSION The phosphate group on the adenine-linked ribose moiety of adenine nucleotides and pyridine nucleotides is presumed to be essential for the observed effect on aggregation. Efficient aggregation requires the presence of the four octarepeats. These insights may be helpful in the eventual development of therapeutic agents against prion-related disorders.
Collapse
Affiliation(s)
- Noriyuki Shiraishi
- Department of Nutrition, Tokai Gakuen University, 2-901 Nakahira, Nagoya 468-8514, Japan
| | - Yoshiaki Hirano
- Department of Nutrition, Tokai Gakuen University, 2-901 Nakahira, Nagoya 468-8514, Japan
| |
Collapse
|
10
|
Restelli E, Capone V, Pozzoli M, Ortolan D, Quaglio E, Corbelli A, Fiordaliso F, Beznoussenko GV, Artuso V, Roiter I, Sallese M, Chiesa R. Activation of Src family kinase ameliorates secretory trafficking in mutant prion protein cells. J Biol Chem 2021; 296:100490. [PMID: 33662396 PMCID: PMC8059059 DOI: 10.1016/j.jbc.2021.100490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 02/04/2021] [Accepted: 02/26/2021] [Indexed: 11/25/2022] Open
Abstract
Fatal familial insomnia (FFI), genetic Creutzfeldt-Jakob disease (gCJD), and Gerstmann-Sträussler-Scheinker (GSS) syndrome are neurodegenerative disorders linked to prion protein (PrP) mutations. The pathogenic mechanisms are not known, but increasing evidence points to mutant PrP misfolding and retention in the secretory pathway. We previously found that the D178N/M129 mutation associated with FFI accumulates in the Golgi of neuronal cells, impairing post-Golgi trafficking. In this study we further characterized the trafficking defect induced by the FFI mutation and tested the 178N/V129 variant linked to gCJD and a nine-octapeptide repeat insertion associated with GSS. We used transfected HeLa cells, embryonic fibroblasts and primary neurons from transgenic mice, and fibroblasts from carriers of the FFI mutation. In all these cell types, the mutant PrPs showed abnormal intracellular localizations, accumulating in the endoplasmic reticulum (ER) and Golgi. To test the efficiency of the membrane trafficking system, we monitored the intracellular transport of the temperature-sensitive vesicular stomatite virus glycoprotein (VSV-G), a well-established cargo reporter, and of endogenous procollagen I (PC-I). We observed marked alterations in secretory trafficking, with VSV-G accumulating mainly in the Golgi complex and PC-I in the ER and Golgi. A redacted version of mutant PrP with reduced propensity to misfold did not impair VSV-G trafficking, nor did artificial ER or Golgi retention of wild-type PrP; this indicates that both misfolding and intracellular retention were required to induce the transport defect. Pharmacological activation of Src family kinase (SFK) improved intracellular transport, suggesting that mutant PrP impairs secretory trafficking through corruption of SFK-mediated signaling.
Collapse
Affiliation(s)
- Elena Restelli
- Laboratory of Prion Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Vanessa Capone
- Department of Innovative Technologies in Medicine & Dentistry, University G. D'Annunzio, Chieti, Italy; Center for Advanced Studies and Technology (CAST), University G. D'Annunzio, Chieti, Italy
| | - Manuela Pozzoli
- Laboratory of Prion Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Davide Ortolan
- Laboratory of Prion Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elena Quaglio
- Laboratory of Prion Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Alessandro Corbelli
- Bio-Imaging Unit, Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Fabio Fiordaliso
- Bio-Imaging Unit, Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | - Ignazio Roiter
- ULSS 2 Marca Trevigiana, Ca' Foncello Hospital, Treviso, Italy
| | - Michele Sallese
- Department of Innovative Technologies in Medicine & Dentistry, University G. D'Annunzio, Chieti, Italy; Center for Advanced Studies and Technology (CAST), University G. D'Annunzio, Chieti, Italy
| | - Roberto Chiesa
- Laboratory of Prion Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| |
Collapse
|
11
|
Marín-Moreno A, Espinosa JC, Torres JM. Transgenic mouse models for the study of prion diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:147-177. [PMID: 32958231 DOI: 10.1016/bs.pmbts.2020.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Prions are unique agents that challenge the molecular biology dogma by transmitting information on the protein level. They cause neurodegenerative diseases that lack of any cure or treatment called transmissible spongiform encephalopathies. The function of the normal form of the prion protein, the exact mechanism of prion propagation between species as well as at the cellular level and neuron degeneration remains elusive. However, great amount of information known for all these aspects has been achieved thanks to the use of animal models and more precisely to transgenic mouse models. In this chapter, the main contributions of these powerful research tools in the prion field are revised.
Collapse
Affiliation(s)
- Alba Marín-Moreno
- Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Spain
| | | | - Juan María Torres
- Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Spain.
| |
Collapse
|
12
|
Hara H, Sakaguchi S. N-Terminal Regions of Prion Protein: Functions and Roles in Prion Diseases. Int J Mol Sci 2020; 21:ijms21176233. [PMID: 32872280 PMCID: PMC7504422 DOI: 10.3390/ijms21176233] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 01/30/2023] Open
Abstract
The normal cellular isoform of prion protein, designated PrPC, is constitutively converted to the abnormally folded, amyloidogenic isoform, PrPSc, in prion diseases, which include Creutzfeldt-Jakob disease in humans and scrapie and bovine spongiform encephalopathy in animals. PrPC is a membrane glycoprotein consisting of the non-structural N-terminal domain and the globular C-terminal domain. During conversion of PrPC to PrPSc, its 2/3 C-terminal region undergoes marked structural changes, forming a protease-resistant structure. In contrast, the N-terminal region remains protease-sensitive in PrPSc. Reverse genetic studies using reconstituted PrPC-knockout mice with various mutant PrP molecules have revealed that the N-terminal domain has an important role in the normal function of PrPC and the conversion of PrPC to PrPSc. The N-terminal domain includes various characteristic regions, such as the positively charged residue-rich polybasic region, the octapeptide repeat (OR) region consisting of five repeats of an octapeptide sequence, and the post-OR region with another positively charged residue-rich polybasic region followed by a stretch of hydrophobic residues. We discuss the normal functions of PrPC, the conversion of PrPC to PrPSc, and the neurotoxicity of PrPSc by focusing on the roles of the N-terminal regions in these topics.
Collapse
|
13
|
Mutant prion proteins increase calcium permeability of AMPA receptors, exacerbating excitotoxicity. PLoS Pathog 2020; 16:e1008654. [PMID: 32673372 PMCID: PMC7365390 DOI: 10.1371/journal.ppat.1008654] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 05/26/2020] [Indexed: 01/26/2023] Open
Abstract
Prion protein (PrP) mutations are linked to genetic prion diseases, a class of phenotypically heterogeneous neurodegenerative disorders with invariably fatal outcome. How mutant PrP triggers neurodegeneration is not known. Synaptic dysfunction precedes neuronal loss but it is not clear whether, and through which mechanisms, disruption of synaptic activity ultimately leads to neuronal death. Here we show that mutant PrP impairs the secretory trafficking of AMPA receptors (AMPARs). Specifically, intracellular retention of the GluA2 subunit results in synaptic exposure of GluA2-lacking, calcium-permeable AMPARs, leading to increased calcium permeability and enhanced sensitivity to excitotoxic cell death. Mutant PrPs linked to different genetic prion diseases affect AMPAR trafficking and function in different ways. Our findings identify AMPARs as pathogenic targets in genetic prion diseases, and support the involvement of excitotoxicity in neurodegeneration. They also suggest a mechanistic explanation for how different mutant PrPs may cause distinct disease phenotypes. Genetic prion diseases are degenerative brain disorders caused by mutations in the gene encoding the prion protein (PrP). Different PrP mutations cause different diseases, including Creutzfeldt-Jakob disease, fatal familial insomnia and Gerstmann-Sträussler-Scheinker syndrome. How mutant PrP causes neuronal death and how different mutants encode distinct disease phenotypes is not known. Here we show that mutant PrP alters the subunit composition of glutamate AMPA receptors, promoting cell surface exposure of GluA2-lacking, calcium-permeable receptors, ultimately increasing neuronal vulnerability to excitotoxic cell death. We also demonstrate that the underlying molecular mechanism is the formation of a GluA2 subunit-PrP complex which is retained in the neuronal secretory pathway. PrP mutants associated with clinically different genetic prion diseases have distinct effects on GluA2 trafficking, depending on their tendency to misfold and aggregate in different intracellular organelles, indicating a possible contribution of this mechanism to the disease phenotype.
Collapse
|
14
|
Melatonin regulates mitochondrial dynamics and alleviates neuron damage in prion diseases. Aging (Albany NY) 2020; 12:11139-11151. [PMID: 32526704 PMCID: PMC7346071 DOI: 10.18632/aging.103328] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/17/2020] [Indexed: 12/30/2022]
Abstract
Prion diseases are neurodegenerative diseases associated with neuron damage and behavioral disorders in animals and humans. Melatonin is a potent antioxidant and is used to treat a variety of diseases. We investigated the neuroprotective effect of melatonin on prion-induced damage in N2a cells. N2a cells were pretreated with 10 μM melatonin for 1 hour followed by incubation with 100 μM PrP106-126 for 24 hours. Melatonin markedly alleviated PrP106-126-induced apoptosis of N2a cells, and inhibited PrP106-126-induced mitochondrial abnormality and dysfunction, including mitochondrial fragmentation and overproduction of reactive oxygen species (ROS), suppression of ATP, reduced mitochondrial membrane potential (MMP), and altered mitochondrial dynamic proteins dynamin-related protein 1 (DRP1) and optic atrophy protein 1 (OPA1). Our findings identify that pretreatment with melatonin prevents the deleterious effects of PrPSc on mitochondrial function and dynamics, protects synapses and alleviates neuron damage. Melatonin could be a novel and effective medication in the therapy of prion diseases.
Collapse
|
15
|
Caughey B, Kraus A. Transmissibility versus Pathogenicity of Self-Propagating Protein Aggregates. Viruses 2019; 11:E1044. [PMID: 31717531 PMCID: PMC6893620 DOI: 10.3390/v11111044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/22/2022] Open
Abstract
The prion-like spreading and accumulation of specific protein aggregates appear to be central to the pathogenesis of many human diseases, including Alzheimer's and Parkinson's. Accumulating evidence indicates that inoculation of tissue extracts from diseased individuals into suitable experimental animals can in many cases induce the aggregation of the disease-associated protein, as well as related pathological lesions. These findings, together with the history of the prion field, have raised the questions about whether such disease-associated protein aggregates are transmissible between humans by casual or iatrogenic routes, and, if so, do they propagate enough in the new host to cause disease? These practical considerations are important because real, and perhaps even only imagined, risks of human-to-human transmission of diseases such as Alzheimer's and Parkinson's may force costly changes in clinical practice that, in turn, are likely to have unintended consequences. The prion field has taught us that a single protein, PrP, can aggregate into forms that can propagate exponentially in vitro, but range from being innocuous to deadly when injected into experimental animals in ways that depend strongly on factors such as conformational subtleties, routes of inoculation, and host responses. In assessing the hazards posed by various disease-associated, self-propagating protein aggregates, it is imperative to consider both their actual transmissibilities and the pathological consequences of their propagation, if any, in recipient hosts.
Collapse
Affiliation(s)
- Byron Caughey
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Allison Kraus
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
16
|
Forloni G, Chiesa R, Bugiani O, Salmona M, Tagliavini F. Review: PrP 106-126 - 25 years after. Neuropathol Appl Neurobiol 2019; 45:430-440. [PMID: 30635947 DOI: 10.1111/nan.12538] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022]
Abstract
A quarter of a century ago, we proposed an innovative approach to study the pathogenesis of prion disease, one of the most intriguing biomedical problems that remains unresolved. The synthesis of a peptide homologous to residues 106-126 of the human prion protein (PrP106-126), a sequence present in the PrP amyloid protein of Gerstmann-Sträussler-Scheinker syndrome patients, provided a tractable tool for investigating the mechanisms of neurotoxicity. Together with several other discoveries at the beginning of the 1990s, PrP106-126 contributed to underpin the role of amyloid in the pathogenesis of protein-misfolding neurodegenerative disorders. Later, the role of oligomers on one hand and of prion-like spreading of pathology on the other further clarified mechanisms shared by different neurodegenerative conditions. Our original report on PrP106-126 neurotoxicity also highlighted a role for programmed cell death in CNS diseases. In this review, we analyse the prion research context in which PrP106-126 first appeared and the advances in our understanding of prion disease pathogenesis and therapeutic perspectives 25 years later.
Collapse
Affiliation(s)
- G Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - R Chiesa
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - O Bugiani
- Department of Biochemistry, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - M Salmona
- Department of Biochemistry, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - F Tagliavini
- Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milano, Italy
| |
Collapse
|
17
|
Agbowuro AA, Huston WM, Gamble AB, Tyndall JDA. Proteases and protease inhibitors in infectious diseases. Med Res Rev 2017; 38:1295-1331. [PMID: 29149530 DOI: 10.1002/med.21475] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/10/2017] [Accepted: 10/17/2017] [Indexed: 12/31/2022]
Abstract
There are numerous proteases of pathogenic organisms that are currently targeted for therapeutic intervention along with many that are seen as potential drug targets. This review discusses the chemical and biological makeup of some key druggable proteases expressed by the five major classes of disease causing agents, namely bacteria, viruses, fungi, eukaryotes, and prions. While a few of these enzymes including HIV protease and HCV NS3-4A protease have been targeted to a clinically useful level, a number are yet to yield any clinical outcomes in terms of antimicrobial therapy. A significant aspect of this review discusses the chemical and pharmacological characteristics of inhibitors of the various proteases discussed. A total of 25 inhibitors have been considered potent and safe enough to be trialed in humans and are at different levels of clinical application. We assess the mechanism of action and clinical performance of the protease inhibitors against infectious agents with their developmental strategies and look to the next frontiers in the use of protease inhibitors as anti-infective agents.
Collapse
Affiliation(s)
| | - Wilhelmina M Huston
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, Australia
| | - Allan B Gamble
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
18
|
Puig B, Altmeppen HC, Glatzel M. Misfolding leads the way to unraveling signaling pathways in the pathophysiology of prion diseases. Prion 2017; 10:434-443. [PMID: 27870599 DOI: 10.1080/19336896.2016.1244593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A misfolded version of the prion protein represents an essential component in the pathophysiology of fatal neurodegenerative prion diseases, which affect humans and animals alike. They may be of sporadic origin, acquired through exogenous introduction of infectious misfolded prion protein, or caused by genetic alterations in the prion protein coding gene. We have recently described a novel pathway linking retention of mutant prion protein in the early secretory pathway to activation p38-MAPK and a neurodegenerative phenotype in transgenic mice. Here we review the consequences that mutations in prion protein have on intracellular transport and stress responses focusing on protein quality control. We also discuss the neurotoxic signaling elicited by the accumulation of mutant prion protein in the endoplasmic reticulum and the Golgi apparatus. Improved knowledge about these processes will help us to better understand complex pathogenesis of prion diseases, a prerequisite for therapeutic strategies.
Collapse
Affiliation(s)
- Berta Puig
- a Institute of Neuropathology, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Hermann C Altmeppen
- a Institute of Neuropathology, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Markus Glatzel
- a Institute of Neuropathology, University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| |
Collapse
|
19
|
Watts JC, Prusiner SB. Experimental Models of Inherited PrP Prion Diseases. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a027151. [PMID: 28096244 DOI: 10.1101/cshperspect.a027151] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The inherited prion protein (PrP) prion disorders, which include familial Creutzfeldt-Jakob disease, Gerstmann-Sträussler-Scheinker disease, and fatal familial insomnia, constitute ∼10%-15% of all PrP prion disease cases in humans. Attempts to generate animal models of these disorders using transgenic mice expressing mutant PrP have produced variable results. Although many lines of mice develop spontaneous signs of neurological illness with accompanying prion disease-specific neuropathological changes, others do not. Furthermore, demonstrating the presence of protease-resistant PrP species and prion infectivity-two of the hallmarks of the PrP prion disorders-in the brains of spontaneously sick mice has proven particularly challenging. Here, we review the progress that has been made toward developing accurate mouse models of the inherited PrP prion disorders.
Collapse
Affiliation(s)
- Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Biochemistry, University of Toronto, Toronto, Ontario M5T 2S8, Canada
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, Departments of Neurology and Biochemistry and Biophysics, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
20
|
Moreno JA, Telling GC. Insights into Mechanisms of Transmission and Pathogenesis from Transgenic Mouse Models of Prion Diseases. Methods Mol Biol 2017; 1658:219-252. [PMID: 28861793 DOI: 10.1007/978-1-4939-7244-9_16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Prions represent a new paradigm of protein-mediated information transfer. In the case of mammals, prions are the cause of fatal, transmissible neurodegenerative diseases, sometimes referred to as transmissible spongiform encephalopathies (TSEs), which frequently occur as epidemics. An increasing body of evidence indicates that the canonical mechanism of conformational corruption of cellular prion protein (PrPC) by the pathogenic isoform (PrPSc) that is the basis of prion formation in TSEs is common to a spectrum of proteins associated with various additional human neurodegenerative disorders, including the more common Alzheimer's and Parkinson's diseases. The peerless infectious properties of TSE prions, and the unparalleled tools for their study, therefore enable elucidation of mechanisms of template-mediated conformational propagation that are generally applicable to these related disease states. Many unresolved issues remain including the exact molecular nature of the prion, the detailed cellular and molecular mechanisms of prion propagation, and the means by which prion diseases can be both genetic and infectious. In addition, we know little about the mechanism by which neurons degenerate during prion diseases. Tied to this, the physiological role of the normal form of the prion protein remains unclear and it is uncertain whether or not loss of this function contributes to prion pathogenesis. The factors governing the transmission of prions between species remain unclear, in particular the means by which prion strains and PrP primary structure interact to affect interspecies prion transmission. Despite all these unknowns, advances in our understanding of prions have occurred because of their transmissibility to experimental animals, and the development of transgenic (Tg) mouse models has done much to further our understanding about various aspects of prion biology. In this review, we will focus on advances in our understanding of prion biology that occurred in the past 8 years since our last review of this topic.
Collapse
Affiliation(s)
- Julie A Moreno
- Cell and Molecular Biology Graduate Program, Molecular, Cellular and Integrative Neuroscience Graduate Program, Department of Microbiology, Immunology and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, 80523, USA
| | - Glenn C Telling
- Cell and Molecular Biology Graduate Program, Molecular, Cellular and Integrative Neuroscience Graduate Program, Department of Microbiology, Immunology and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, 80523, USA.
| |
Collapse
|
21
|
Barron RM, King D, Jeffrey M, McGovern G, Agarwal S, Gill AC, Piccardo P. PrP aggregation can be seeded by pre-formed recombinant PrP amyloid fibrils without the replication of infectious prions. Acta Neuropathol 2016; 132:611-24. [PMID: 27376534 PMCID: PMC5023723 DOI: 10.1007/s00401-016-1594-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/08/2016] [Accepted: 06/26/2016] [Indexed: 12/22/2022]
Abstract
Mammalian prions are unusual infectious agents, as they are thought to consist solely of aggregates of misfolded prion protein (PrP). Generation of synthetic prions, composed of recombinant PrP (recPrP) refolded into fibrils, has been utilised to address whether PrP aggregates are, indeed, infectious prions. In several reports, neurological disease similar to transmissible spongiform encephalopathy (TSE) has been described following inoculation and passage of various forms of fibrils in transgenic mice and hamsters. However, in studies described here, we show that inoculation of recPrP fibrils does not cause TSE disease, but, instead, seeds the formation of PrP amyloid plaques in PrP-P101L knock-in transgenic mice (101LL). Importantly, both WT-recPrP fibrils and 101L-recPrP fibrils can seed plaque formation, indicating that the fibrillar conformation, and not the primary sequence of PrP in the inoculum, is important in initiating seeding. No replication of infectious prions or TSE disease was observed following both primary inoculation and subsequent subpassage. These data, therefore, argue against recPrP fibrils being infectious prions and, instead, indicate that these pre-formed seeds are acting to accelerate the formation of PrP amyloid plaques in 101LL Tg mice. In addition, these data reproduce a phenotype which was previously observed in 101LL mice following inoculation with brain extract containing in vivo-generated PrP amyloid fibrils, which has not been shown for other synthetic prion models. These data are reminiscent of the "prion-like" spread of aggregated forms of the beta-amyloid peptide (Aβ), α-synuclein and tau observed following inoculation of transgenic mice with pre-formed seeds of each misfolded protein. Hence, even when the protein is PrP, misfolding and aggregation do not reproduce the full clinicopathological phenotype of disease. The initiation and spread of protein aggregation in transgenic mouse lines following inoculation with pre-formed fibrils may, therefore, more closely resemble a seeded proteinopathy than an infectious TSE disease.
Collapse
Affiliation(s)
- Rona M Barron
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK.
| | - Declan King
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK
| | - Martin Jeffrey
- Animal and Plant Health Agency, Pentlands Science Park, Midlothian, Scotland, UK
| | - Gillian McGovern
- Animal and Plant Health Agency, Pentlands Science Park, Midlothian, Scotland, UK
| | - Sonya Agarwal
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK
| | - Andrew C Gill
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK
| | - Pedro Piccardo
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK
| |
Collapse
|
22
|
Abstract
Transmissible spongiform encephalopathies (TSEs), or prion diseases, are fatal neurodegenerative disorders characterised by long incubation period, short clinical duration, and transmissibility to susceptible species. Neuronal loss, spongiform changes, gliosis and the accumulation in the brain of the misfolded version of a membrane-bound cellular prion protein (PrP(C)), termed PrP(TSE), are diagnostic markers of these diseases. Compelling evidence links protein misfolding and its accumulation with neurodegenerative changes. Accordingly, several mechanisms of prion-mediated neurotoxicity have been proposed. In this paper, we provide an overview of the recent knowledge on the mechanisms of neuropathogenesis, the neurotoxic PrP species and the possible therapeutic approaches to treat these devastating disorders.
Collapse
|
23
|
Herms J, Dorostkar MM. Dendritic Spine Pathology in Neurodegenerative Diseases. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 11:221-50. [PMID: 26907528 DOI: 10.1146/annurev-pathol-012615-044216] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Substantial progress has been made toward understanding the neuropathology, genetic origins, and epidemiology of neurodegenerative diseases, including Alzheimer's disease; tauopathies, such as frontotemporal dementia; α-synucleinopathies, such as Parkinson's disease or dementia with Lewy bodies; Huntington's disease; and amyotrophic lateral sclerosis with dementia, as well as prion diseases. Recent evidence has implicated dendritic spine dysfunction as an important substrate of the pathogenesis of dementia in these disorders. Dendritic spines are specialized structures, extending from the neuronal processes, on which excitatory synaptic contacts are formed, and the loss of dendritic spines correlates with the loss of synaptic function. We review the literature that has implicated direct or indirect structural alterations at dendritic spines in the pathogenesis of major neurodegenerative diseases, focusing on those that lead to dementias such as Alzheimer's, Parkinson's, and Huntington's diseases, as well as frontotemporal dementia and prion diseases. We stress the importance of in vivo studies in animal models.
Collapse
Affiliation(s)
- Jochen Herms
- Center for Neuropathology and Prion Research, Ludwig Maximilian University, 81377 Munich, Germany; .,Munich Cluster for Systems Neurology, Ludwig Maximilian University, 81377 Munich, Germany.,German Center for Neurodegenerative Diseases, 81377 Munich, Germany
| | - Mario M Dorostkar
- Center for Neuropathology and Prion Research, Ludwig Maximilian University, 81377 Munich, Germany;
| |
Collapse
|
24
|
Liu C, Li X, Lu B. The Immp2l mutation causes age-dependent degeneration of cerebellar granule neurons prevented by antioxidant treatment. Aging Cell 2016; 15:167-76. [PMID: 26616244 PMCID: PMC4717271 DOI: 10.1111/acel.12426] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2015] [Indexed: 12/04/2022] Open
Abstract
Reactive oxygen species are implicated in age‐associated neurodegeneration, although direct in vivo evidence is lacking. We recently showed that mice with a mutation in the Inner Mitochondrial Membrane Peptidase 2‐like (Immp2l) gene had elevated levels of mitochondrial superoxide, impaired fertility and age‐associated phenotypes, including kyphosis and ataxia. Here we show that ataxia and cerebellar hypoplasia occur in old mutant mice (> 16 months). Cerebellar granule neurons (CGNs) are significantly underrepresented; Purkinje cells and cells in the molecular layer are not affected. Treating mutant mice with the mitochondria‐targeted antioxidant SkQ1 from 6 weeks to 21 months protected cerebellar granule neurons. Apoptotic granule neurons were observed in mutant mice but not in age‐matched normal control mice or SkQ1‐treated mice. Old mutant mice showed increased serum protein carbonyl content, cerebellar 4‐hydroxynonenal (HNE), and nitrotyrosine modification compared to old normal control mice. SOD2 expression was increased in Purkinje cells but decreased in granule neurons of old mutant mice. Mitochondrial marker protein VDAC1 also was decreased in CGNs of old mutant mice, suggesting decreased mitochondrial number. SkQ1 treatment decreased HNE and nitrotyrosine modification, and restored SOD2 and VDAC1 expression in CGNs of old mutant mice. Neuronal expression of nitric oxide synthase was increased in cerebella of young mutant mice but decreased in old mutant mice. Our work provides evidence for a causal role of oxidative stress in neurodegeneration of Immp2l mutant mice. The Immp2l mutant mouse model could be valuable in elucidating the role of oxidative stress in age‐associated neurodegeneration.
Collapse
Affiliation(s)
- Chunlian Liu
- Department of Center for Reproductive Medicine General Hospital Ningxia Medical University Ningxia 750004 China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education Ningxia Medical University Ningxia 750004 China
- Wake Forest University Health Sciences Institute for Regenerative Medicine Winston‐Salem NC 27157 USA
| | - Xue Li
- Wake Forest University Health Sciences Institute for Regenerative Medicine Winston‐Salem NC 27157 USA
- Department of Pathology Beijing Chao‐Yang Hospital Capital Medical University Beijing China
| | - Baisong Lu
- Wake Forest University Health Sciences Institute for Regenerative Medicine Winston‐Salem NC 27157 USA
| |
Collapse
|
25
|
Veber D, Scalabrino G. Are PrPCs involved in some human myelin diseases? Relating experimental studies to human pathology. J Neurol Sci 2015; 359:396-403. [DOI: 10.1016/j.jns.2015.09.365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/04/2015] [Accepted: 09/23/2015] [Indexed: 11/29/2022]
|
26
|
Bouybayoune I, Mantovani S, Del Gallo F, Bertani I, Restelli E, Comerio L, Tapella L, Baracchi F, Fernández-Borges N, Mangieri M, Bisighini C, Beznoussenko GV, Paladini A, Balducci C, Micotti E, Forloni G, Castilla J, Fiordaliso F, Tagliavini F, Imeri L, Chiesa R. Transgenic fatal familial insomnia mice indicate prion infectivity-independent mechanisms of pathogenesis and phenotypic expression of disease. PLoS Pathog 2015; 11:e1004796. [PMID: 25880443 PMCID: PMC4400166 DOI: 10.1371/journal.ppat.1004796] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 03/09/2015] [Indexed: 11/18/2022] Open
Abstract
Fatal familial insomnia (FFI) and a genetic form of Creutzfeldt-Jakob disease (CJD178) are clinically different prion disorders linked to the D178N prion protein (PrP) mutation. The disease phenotype is determined by the 129 M/V polymorphism on the mutant allele, which is thought to influence D178N PrP misfolding, leading to the formation of distinctive prion strains with specific neurotoxic properties. However, the mechanism by which misfolded variants of mutant PrP cause different diseases is not known. We generated transgenic (Tg) mice expressing the mouse PrP homolog of the FFI mutation. These mice synthesize a misfolded form of mutant PrP in their brains and develop a neurological illness with severe sleep disruption, highly reminiscent of FFI and different from that of analogously generated Tg(CJD) mice modeling CJD178. No prion infectivity was detectable in Tg(FFI) and Tg(CJD) brains by bioassay or protein misfolding cyclic amplification, indicating that mutant PrP has disease-encoding properties that do not depend on its ability to propagate its misfolded conformation. Tg(FFI) and Tg(CJD) neurons have different patterns of intracellular PrP accumulation associated with distinct morphological abnormalities of the endoplasmic reticulum and Golgi, suggesting that mutation-specific alterations of secretory transport may contribute to the disease phenotype. Genetic prion diseases are degenerative brain disorders caused by mutations in the gene encoding the prion protein (PrP). Different PrP mutations cause different diseases, including Creutzfeldt-Jakob disease (CJD) and fatal familial insomnia (FFI). The reason for this variability is not known, but assembly of the mutant PrPs into distinct aggregates that spread in the brain by promoting PrP aggregation may contribute to the disease phenotype. We previously generated transgenic mice modeling genetic CJD, clinically identified by dementia and motor abnormalities. We have now generated transgenic mice carrying the PrP mutation associated with FFI, and found that they develop severe sleep abnormalities and other key features of the human disorder. Thus, transgenic mice recapitulate the phenotypic differences seen in humans. The mutant PrPs in FFI and CJD mice are aggregated but unable to promote PrP aggregation. They accumulate in different intracellular compartments and cause distinct morphological abnormalities of transport organelles. These results indicate that mutant PrP has disease-encoding properties that are independent of its ability to self-propagate, and suggest that the phenotypic heterogeneity may be due to different effects of aggregated PrP on intracellular transport. Our study provides new insights into the mechanisms of selective neuronal dysfunction due to protein aggregation.
Collapse
Affiliation(s)
- Ihssane Bouybayoune
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Susanna Mantovani
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Federico Del Gallo
- Department of Health Sciences, University of Milan Medical School, Milan, Italy
| | - Ilaria Bertani
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Elena Restelli
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Liliana Comerio
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Laura Tapella
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Francesca Baracchi
- Department of Health Sciences, University of Milan Medical School, Milan, Italy
| | | | - Michela Mangieri
- Division of Neuropathology and Neurology, IRCCS Foundation “Carlo Besta” National Neurological Institute, Milan, Italy
| | - Cinzia Bisighini
- Bio-Imaging Unit, Department of Cardiovascular Research, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | | | - Alessandra Paladini
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Claudia Balducci
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Joaquín Castilla
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Fabio Fiordaliso
- Bio-Imaging Unit, Department of Cardiovascular Research, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Fabrizio Tagliavini
- Division of Neuropathology and Neurology, IRCCS Foundation “Carlo Besta” National Neurological Institute, Milan, Italy
| | - Luca Imeri
- Department of Health Sciences, University of Milan Medical School, Milan, Italy
| | - Roberto Chiesa
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
- * E-mail:
| |
Collapse
|
27
|
Scalabrino G, Veber D, Tredici G. Relationships between cobalamin, epidermal growth factor, and normal prions in the myelin maintenance of central nervous system. Int J Biochem Cell Biol 2014; 55:232-41. [PMID: 25239885 DOI: 10.1016/j.biocel.2014.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/06/2014] [Accepted: 09/08/2014] [Indexed: 01/08/2023]
Abstract
Cobalamin (Cbl), epidermal growth factor (EGF), and prions (PrPs) are key molecules for myelin maintenance in the central and peripheral nervous systems. Cbl and EGF increase normal prion (PrP(C)) synthesis and PrP(C) levels in rat spinal cord (SC) and elsewhere. Cbl deficiency increases PrP(C) levels in rat SC and cerebrospinal fluid (CSF), and decreases PrP(C)-mRNA levels in rat SC. The administration of anti-octapeptide repeat PrP(C) region antibodies (Abs) to Cbl-deficient (Cbl-D) rats prevents SC myelin lesions and a local increase in tumor necrosis factor (TNF)-α levels, whereas anti-TNF-α Abs prevent SC myelin lesions and the increase in SC and CSF PrP(C) levels. As it is known that both Cbl and EGF regulate SC PrP(C) synthesis independently, and that Cbl regulates SC EGF synthesis, EGF may play both Cbl-independent and Cbl-dependent roles. When Cbl-D rats undergo Cbl replacement therapy, SC PrP(C) levels are similar to those observed in Cbl-D rats. In rat frontal cortex (which is marginally affected by Cbl deficiency in histological terms), Cbl deficiency decreases PrP(C) levels and the increase induced by Cbl replacement leads to their normalization. Increased nerve PrP(C) levels are detected in the myelin lesions of the peripheral neuropathy of Cbl-D rats, and CSF PrP(C) levels are also increased in Cbl-D patients (but not in patients with Cbl-unrelated neurological diseases). Various common steps in the downstream signaling pathway of Cbl, EGF, and PrP(C) underlines the close relationship between the three molecules in keeping myelin normal.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences, Laboratory of Neuropathology, University of Milan, 20133 Milano, Italy.
| | - Daniela Veber
- Department of Biomedical Sciences, Laboratory of Neuropathology, University of Milan, 20133 Milano, Italy
| | - Giovanni Tredici
- Department of Translational Medicine and Surgery, University of Milano-Bicocca, 20052 Monza, Italy
| |
Collapse
|
28
|
Torres JM, Castilla J, Pintado B, Gutiérrez-Adan A, Andréoletti O, Aguilar-Calvo P, Arroba AI, Parra-Arrondo B, Ferrer I, Manzanares J, Espinosa JC. Spontaneous generation of infectious prion disease in transgenic mice. Emerg Infect Dis 2014; 19:1938-47. [PMID: 24274622 PMCID: PMC3840888 DOI: 10.3201/eid1912.130106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We generated transgenic mice expressing bovine cellular prion protein (PrPC) with a leucine substitution at codon 113 (113L). This protein is homologous to human protein with mutation 102L, and its genetic link with Gerstmann–Sträussler–Scheinker syndrome has been established. This mutation in bovine PrPC causes a fully penetrant, lethal, spongiform encephalopathy. This genetic disease was transmitted by intracerebral inoculation of brain homogenate from ill mice expressing mutant bovine PrP to mice expressing wild-type bovine PrP, which indicated de novo generation of infectious prions. Our findings demonstrate that a single amino acid change in the PrPC sequence can induce spontaneous generation of an infectious prion disease that differs from all others identified in hosts expressing the same PrPC sequence. These observations support the view that a variety of infectious prion strains might spontaneously emerge in hosts displaying random genetic PrPC mutations.
Collapse
|
29
|
Davidson L, Knight R. Neuropathogenesis of prion disease. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.13.74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT: Although much is known about prion diseases (characterized by a post-translational misfolding of the prion protein [PrP]) and their neuropathology and molecular pathology, the fundamental cause of illness, the basic neuropathogenesis, remains uncertain. There are three broad considerations discussed in this review: the possible loss of normal PrP function, the possible direct toxicity of the abnormally folded PrP and a harmful interaction between the normal and abnormal protein. In considering these possibilities, there are difficulties, including the facts that the relevant normal functions of the PrP are somewhat uncertain and that there are a number of possible toxic species of abnormal protein. In addition to the possible interactions of normal and abnormal PrP in prion disease, PrP may play a role in the neuropathogenesis of other diseases (such as Alzheimer’s disease).
Collapse
Affiliation(s)
- Louise Davidson
- National Creutzfeldt–Jakob Disease Research & Surveillance Unit, University of Edinburgh, Edinburgh, UK
| | - Richard Knight
- National Creutzfeldt–Jakob Disease Research & Surveillance Unit, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
30
|
Asuni AA, Gray B, Bailey J, Skipp P, Perry VH, O'Connor V. Analysis of the hippocampal proteome in ME7 prion disease reveals a predominant astrocytic signature and highlights the brain-restricted production of clusterin in chronic neurodegeneration. J Biol Chem 2013; 289:4532-45. [PMID: 24366862 PMCID: PMC3924314 DOI: 10.1074/jbc.m113.502690] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prion diseases are characterized by accumulation of misfolded protein, gliosis, synaptic dysfunction, and ultimately neuronal loss. This sequence, mirroring key features of Alzheimer disease, is modeled well in ME7 prion disease. We used iTRAQTM/mass spectrometry to compare the hippocampal proteome in control and late-stage ME7 animals. The observed changes associated with reactive glia highlighted some specific proteins that dominate the proteome in late-stage disease. Four of the up-regulated proteins (GFAP, high affinity glutamate transporter (EAAT-2), apo-J (Clusterin), and peroxiredoxin-6) are selectively expressed in astrocytes, but astrocyte proliferation does not contribute to their up-regulation. The known functional role of these proteins suggests this response acts against protein misfolding, excitotoxicity, and neurotoxic reactive oxygen species. A recent convergence of genome-wide association studies and the peripheral measurement of circulating levels of acute phase proteins have focused attention on Clusterin as a modifier of late-stage Alzheimer disease and a biomarker for advanced neurodegeneration. Since ME7 animals allow independent measurement of acute phase proteins in the brain and circulation, we extended our investigation to address whether changes in the brain proteome are detectable in blood. We found no difference in the circulating levels of Clusterin in late-stage prion disease when animals will show behavioral decline, accumulation of misfolded protein, and dramatic synaptic and neuronal loss. This does not preclude an important role of Clusterin in late-stage disease, but it cautions against the assumption that brain levels provide a surrogate peripheral measure for the progression of brain degeneration.
Collapse
|
31
|
Ashrafuzzaman M, Tseng CY, Kapty J, Mercer JR, Tuszynski JA. A computationally designed DNA aptamer template with specific binding to phosphatidylserine. Nucleic Acid Ther 2013; 23:418-26. [PMID: 24279298 PMCID: PMC3868250 DOI: 10.1089/nat.2013.0415] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 10/28/2013] [Indexed: 11/12/2022] Open
Abstract
The phospholipid phosphatidylserine (PS) is an early marker exploited for detecting apoptosis (PS externalization in the cell membrane bilayer) and one factor that is associated with increased amyloid plaque deposition in transmissible spongiform encephalopathies (TSEs). PS can therefore be considered as a promising target for diagnosis or treatment of diseases. Aptamers (short nucleic acid sequences) are a particularly attractive class of materials among those currently considered for targeting PS. Here we applied an entropy based seed-and-grow strategy to design a DNA aptamer template to bind specifically to PS. The binding properties of designed aptamers were investigated computationally and experimentally. The studies identify the sequence, 5'-AAAGAC-3', as the preferred template for further modifications and studies toward its practical implementations.
Collapse
Affiliation(s)
- Md Ashrafuzzaman
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Chih-Yuan Tseng
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Janice Kapty
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John R. Mercer
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jack A. Tuszynski
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
32
|
Synaptic dysfunction in prion diseases: a trafficking problem? Int J Cell Biol 2013; 2013:543803. [PMID: 24369467 PMCID: PMC3863542 DOI: 10.1155/2013/543803] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/08/2013] [Indexed: 11/26/2022] Open
Abstract
Synaptic dysfunction is an important cause of neurological symptoms in prion diseases, a class of clinically heterogeneous neurodegenerative disorders caused by misfolding of the cellular prion protein (PrPC). Experimental data suggest that accumulation of misfolded PrPC in the endoplasmic reticulum (ER) may be crucial in synaptic failure, possibly because of the activation of the translational repression pathway of the unfolded protein response. Here, we report that this pathway is not operative in mouse models of genetic prion disease, consistent with our previous observation that ER stress is not involved. Building on our recent finding that ER retention of mutant PrPC impairs the secretory trafficking of calcium channels essential for synaptic function, we propose a model of pathogenicity in which intracellular retention of misfolded PrPC results in loss of function or gain of toxicity of PrPC-interacting proteins. This neurotoxic modality may also explain the phenotypic heterogeneity of prion diseases.
Collapse
|
33
|
Myelin damage due to local quantitative abnormalities in normal prion levels: evidence from subacute combined degeneration and multiple sclerosis. J Neurol 2013; 261:1451-60. [DOI: 10.1007/s00415-013-7152-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/07/2013] [Accepted: 10/08/2013] [Indexed: 10/26/2022]
|
34
|
Abstract
Transmissible encephalopathies (TSEs) are believed by many to arise by spontaneous conversion of host prion protein (PrP) into an infectious amyloid (PrP-res, PrP (Sc) ) without nucleic acid. Many TSE agents reside in the environment, with infection controlled by public health measures. These include the disappearance of kuru with the cessation of ritual cannibalism, the dramatic reduction of epidemic bovine encephalopathy (BSE) by removal of contaminated feed, and the lack of endemic scrapie in geographically isolated Australian sheep with susceptible PrP genotypes. While prion protein modeling has engendered an intense focus on common types of protein misfolding and amyloid formation in diverse organisms and diseases, the biological characteristics of infectious TSE agents, and their recognition by the host as foreign entities, raises several fundamental new directions for fruitful investigation such as: (1) unrecognized microbial agents in the environmental metagenome that may cause latent neurodegenerative disease, (2) the evolutionary social and protective functions of different amyloid proteins in diverse organisms from bacteria to mammals, and (3) amyloid formation as a beneficial innate immune response to stress (infectious and non-infectious). This innate process however, once initiated, can become unstoppable in accelerated neuronal aging.
Collapse
|
35
|
Senatore A, Colleoni S, Verderio C, Restelli E, Morini R, Condliffe S, Bertani I, Mantovani S, Canovi M, Micotti E, Forloni G, Dolphin A, Matteoli M, Gobbi M, Chiesa R. Mutant PrP suppresses glutamatergic neurotransmission in cerebellar granule neurons by impairing membrane delivery of VGCC α(2)δ-1 Subunit. Neuron 2012; 74:300-13. [PMID: 22542184 PMCID: PMC3339322 DOI: 10.1016/j.neuron.2012.02.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2012] [Indexed: 01/17/2023]
Abstract
How mutant prion protein (PrP) leads to neurological dysfunction in genetic prion diseases is unknown. Tg(PG14) mice synthesize a misfolded mutant PrP which is partially retained in the neuronal endoplasmic reticulum (ER). As these mice age, they develop ataxia and massive degeneration of cerebellar granule neurons (CGNs). Here, we report that motor behavioral deficits in Tg(PG14) mice emerge before neurodegeneration and are associated with defective glutamate exocytosis from granule neurons due to impaired calcium dynamics. We found that mutant PrP interacts with the voltage-gated calcium channel α(2)δ-1 subunit, which promotes the anterograde trafficking of the channel. Owing to ER retention of mutant PrP, α(2)δ-1 accumulates intracellularly, impairing delivery of the channel complex to the cell surface. Thus, mutant PrP disrupts cerebellar glutamatergic neurotransmission by reducing the number of functional channels in CGNs. These results link intracellular PrP retention to synaptic dysfunction, indicating new modalities of neurotoxicity and potential therapeutic strategies.
Collapse
Affiliation(s)
- Assunta Senatore
- Dulbecco Telethon Institute, 20156 Milan, Italy, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Simona Colleoni
- Department of Biochemistry and Molecular Pharmacology, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Claudia Verderio
- Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche Institute of Neuroscience, University of Milan, 20129 Milan, Italy
| | - Elena Restelli
- Dulbecco Telethon Institute, 20156 Milan, Italy, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Raffaella Morini
- Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche Institute of Neuroscience, University of Milan, 20129 Milan, Italy
| | - Steven B. Condliffe
- Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche Institute of Neuroscience, University of Milan, 20129 Milan, Italy
| | - Ilaria Bertani
- Dulbecco Telethon Institute, 20156 Milan, Italy, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Susanna Mantovani
- Dulbecco Telethon Institute, 20156 Milan, Italy, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Mara Canovi
- Department of Biochemistry and Molecular Pharmacology, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Annette C. Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, WC1E6BT London, UK
| | - Michela Matteoli
- Department of Medical Pharmacology and Consiglio Nazionale delle Ricerche Institute of Neuroscience, University of Milan, 20129 Milan, Italy
- Istituto Clinico Humanitas IRCCS, 20089 Milan, Italy
| | - Marco Gobbi
- Department of Biochemistry and Molecular Pharmacology, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
| | - Roberto Chiesa
- Dulbecco Telethon Institute, 20156 Milan, Italy, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, 20156 Milan, Italy
- Corresponding author
| |
Collapse
|
36
|
Quaglio E, Restelli E, Garofoli A, Dossena S, De Luigi A, Tagliavacca L, Imperiale D, Migheli A, Salmona M, Sitia R, Forloni G, Chiesa R. Expression of mutant or cytosolic PrP in transgenic mice and cells is not associated with endoplasmic reticulum stress or proteasome dysfunction. PLoS One 2011; 6:e19339. [PMID: 21559407 PMCID: PMC3084828 DOI: 10.1371/journal.pone.0019339] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 03/27/2011] [Indexed: 12/20/2022] Open
Abstract
The cellular pathways activated by mutant prion protein (PrP) in genetic prion diseases, ultimately leading to neuronal dysfunction and degeneration, are not known. Several mutant PrPs misfold in the early secretory pathway and reside longer in the endoplasmic reticulum (ER) possibly stimulating ER stress-related pathogenic mechanisms. To investigate whether mutant PrP induced maladaptive responses, we checked key elements of the unfolded protein response (UPR) in transgenic mice, primary neurons and transfected cells expressing two different mutant PrPs. Because ER stress favors the formation of untranslocated PrP that might aggregate in the cytosol and impair proteasome function, we also measured the activity of the ubiquitin proteasome system (UPS). Molecular, biochemical and immunohistochemical analyses found no increase in the expression of UPR-regulated genes, such as Grp78/Bip, CHOP/GADD153, or ER stress-dependent splicing of the mRNA encoding the X-box-binding protein 1. No alterations in UPS activity were detected in mutant mouse brains and primary neurons using the Ub(G76V)-GFP reporter and a new fluorogenic peptide for monitoring proteasomal proteolytic activity in vivo. Finally, there was no loss of proteasome function in neurons in which endogenous PrP was forced to accumulate in the cytosol by inhibiting cotranslational translocation. These results indicate that neither ER stress, nor perturbation of proteasome activity plays a major pathogenic role in prion diseases.
Collapse
Affiliation(s)
- Elena Quaglio
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Elena Restelli
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Anna Garofoli
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Sara Dossena
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Ada De Luigi
- Department of Biochemistry and Molecular Pharmacology, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Luigina Tagliavacca
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Università Vita-Salute San Raffaele, Milan, Italy
| | - Daniele Imperiale
- Neurology Unit, Human Prion Diseases Center D.O.M.P., Maria Vittoria Hospital, Torino, Italy
| | - Antonio Migheli
- Neurology Unit, Human Prion Diseases Center D.O.M.P., Maria Vittoria Hospital, Torino, Italy
| | - Mario Salmona
- Department of Biochemistry and Molecular Pharmacology, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Roberto Sitia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Università Vita-Salute San Raffaele, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Roberto Chiesa
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| |
Collapse
|
37
|
Prion protein and its conformational conversion: a structural perspective. Top Curr Chem (Cham) 2011; 305:135-67. [PMID: 21630136 DOI: 10.1007/128_2011_165] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The key molecular event in the pathogenesis of prion diseases is the conformational conversion of a cellular prion protein, PrP(C), into a misfolded form, PrP(Sc). In contrast to PrP(C) that is monomeric and α-helical, PrP(Sc) is oligomeric in nature and rich in β-sheet structure. According to the "protein-only" model, PrP(Sc) itself represents the infectious prion agent responsible for transmissibility of prion disorders. While this model is supported by rapidly growing experimental data, detailed mechanistic and structural aspects of prion protein conversion remain enigmatic. In this chapter we describe recent advances in understanding biophysical and biochemical aspects of prion diseases, with a special focus on structural underpinnings of prion protein conversion, the structural basis of prion strains, and generation of prion infectivity in vitro from bacterially-expressed recombinant PrP.
Collapse
|
38
|
Li L, Napper S, Cashman NR. Immunotherapy for prion diseases: opportunities and obstacles. Immunotherapy 2010; 2:269-82. [PMID: 20635933 DOI: 10.2217/imt.10.3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Transmissible spongiform encephalopathies (TSEs) represent a unique form of infectious disease based on the misfolding of a self-protein into a pathological conformation. While other human diseases are also attributed to protein misfolding, the TSEs are unique in their zoonotic potential and iatrogenic infectivity. These characteristics are of particular importance in the aftermath of the UK bovine spongiform encephalopathy (BSE) outbreak due to the dual concerns that a subpopulation of individuals exposed to the infectious agent may be serving as asymptomatic carriers, and that TSEs of other food animals may also threaten human health. These potentials, in addition to the ongoing baseline of familial and sporadic human prion diseases, necessitate development of effective treatment options. While TSEs represent a novel paradigm of infection, there is nevertheless the opportunity to apply traditional approaches of medicine for disease treatment and prevention, including vaccines for immunotherapy and immunoprophylaxis. However, vaccine development for TSEs is complicated by the challenges and potential dangers associated with induction of immune responses to a self-epitope, as well as the obstacles to treatment of a chronic infection through immunotherapy. The ongoing threat of TSEs to human health, together with the opportunity to apply information emerging from these investigations to other protein misfolding disorders, justifies the efforts required to overcome these obstacles.
Collapse
Affiliation(s)
- Li Li
- University of British Columbia & Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
39
|
Abstract
Prions are self-propagating proteinaceous infectious agents capable of transmitting disease in the absence of nucleic acids. The nature of the infectious agent in prion diseases has been at the center of passionate debate for the past 30 years. However, recent reports on the in vitro generation of prions have settled all doubts that the misfolded prion protein (PrP(Sc)) is the key component in propagating infectivity. However, we still do not understand completely the mechanism of prion replication and whether or not other cellular factors besides PrP(Sc) are required for infectivity. In this article, we discuss these recent reports under the context of the protein-only hypothesis and their implications.
Collapse
Affiliation(s)
- Rodrigo Diaz-Espinoza
- Mitchell Center for Alzheimer's disease and related brain disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | | |
Collapse
|
40
|
Abstract
The crucial event in the development of transmissible spongiform encephalopathies (TSEs) is the conformational change of a host-encoded membrane protein - the cellular PrPC - into a disease associated, fibril-forming isoform PrPSc. This conformational transition from the α-helix-rich cellular form into the mainly β-sheet containing counterpart initiates an ‘autocatalytic’ reaction which leads to the accumulation of amyloid fibrils in the central nervous system (CNS) and to neurodegeneration, a hallmark of TSEs. The exact molecular mechanisms which lead to the conformational change are still unknown. It also remains to be brought to light how a polypeptide chain can adopt at least two stable conformations. This review focuses on structural aspects of the prion protein with regard to protein-protein interactions and the initiation of prion protein misfolding. It therefore highlights parts of the protein which might play a notable role in the conformational transition from PrPC to PrPSc and consequently in inducing a fatal chain reaction of protein misfolding. Furthermore, features of different proteins, which are able to adopt insoluble fibrillar states under certain circumstances, are compared to PrP in an attempt to understand the unique characteristics of prion diseases.
Collapse
Affiliation(s)
- L Kupfer
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | | | | |
Collapse
|
41
|
Biasini E, Tapella L, Mantovani S, Stravalaci M, Gobbi M, Harris DA, Chiesa R. Immunopurification of pathological prion protein aggregates. PLoS One 2009; 4:e7816. [PMID: 19915706 PMCID: PMC2773113 DOI: 10.1371/journal.pone.0007816] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Accepted: 10/15/2009] [Indexed: 11/23/2022] Open
Abstract
Background Prion diseases are fatal neurodegenerative disorders that can arise sporadically, be genetically inherited or acquired through infection. The key event in these diseases is misfolding of the cellular prion protein (PrPC) into a pathogenic isoform that is rich in β-sheet structure. This conformational change may result in the formation of PrPSc, the prion isoform of PrP, which propagates itself by imprinting its aberrant conformation onto PrPC molecules. A great deal of effort has been devoted to developing protocols for purifying PrPSc for structural studies, and testing its biological properties. Most procedures rely on protease digestion, allowing efficient purification of PrP27-30, the protease-resistant core of PrPSc. However, protease treatment cannot be used to isolate abnormal forms of PrP lacking conventional protease resistance, such as those found in several genetic and atypical sporadic cases. Principal Findings We developed a method for purifying pathological PrP molecules based on sequential centrifugation and immunoprecipitation with a monoclonal antibody selective for aggregated PrP. With this procedure we purified full-length PrPSc and mutant PrP aggregates at electrophoretic homogeneity. PrPSc purified from prion-infected mice was able to seed misfolding of PrPC in a protein misfolding cyclic amplification reaction, and mutant PrP aggregates from transgenic mice were toxic to cultured neurons. Significance The immunopurification protocol described here isolates biologically active forms of aggregated PrP. These preparations may be useful for investigating the structural and chemico-physical properties of infectious and neurotoxic PrP aggregates.
Collapse
Affiliation(s)
- Emiliano Biasini
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Laura Tapella
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Susanna Mantovani
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Matteo Stravalaci
- Department of Biochemistry and Molecular Pharmacology, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Marco Gobbi
- Department of Biochemistry and Molecular Pharmacology, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - David A. Harris
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Roberto Chiesa
- Dulbecco Telethon Institute, Milan, Italy
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
- * E-mail:
| |
Collapse
|
42
|
Abstract
Prions are responsible for a heterogeneous group of fatal neurodegenerative diseases. They can be sporadic, genetic, or infectious disorders involving post-translational modifications of the cellular prion protein (PrP(C)). Prions (PrP(Sc)) are characterized by their infectious property and intrinsic ability to convert the physiological PrP(C) into the pathological form, acting as a template. The "protein-only" hypothesis, postulated by Stanley B. Prusiner, implies the possibility to generate de novo prions in vivo and in vitro. Here we describe major milestones towards proving this hypothesis, taking into account physiological environment/s, biochemical properties and interactors of the PrP(C).
Collapse
Affiliation(s)
- Federico Benetti
- Laboratory of Prion Biology, Neurobiology Sector, Scuola Internazionale Superiore di Studi Avanzati-International School of Advanced Studies (SISSA-ISAS), Basovizza (TS), Italy
| | | |
Collapse
|
43
|
Solomon IH, Schepker JA, Harris DA. Prion neurotoxicity: insights from prion protein mutants. Curr Issues Mol Biol 2009; 12:51-61. [PMID: 19767650 PMCID: PMC4821541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023] Open
Abstract
The chemical nature of prions and the mechanism by which they propagate are now reasonably well understood. In contrast, much less is known about the identity of the toxic prion protein (PrP) species that are responsible for neuronal death, and the cellular pathways that these forms activate. In addition, the normal, physiological function of cellular PrP (PrP(C)) has remained mysterious, hampering efforts to determine whether loss or alteration of this function contributes to the disease phenotype. Considerable evidence now suggests that aggregation, toxicity, and infectivity are distinct properties of PrP that do no necessarily coincide. In this review, we will discuss several mutant forms of PrP that produce spontaneous neurodegeneration in humans and/or transgenic mice without the formation of infectious PrP(Sc). These include an octapeptide insertional mutation, point mutations that favor synthesis of transmembrane forms of PrP, and deletions encompassing the central domain whose neurotoxicity is antagonized by the presence of wild-type PrP. By isolating the neurotoxic effects of PrP from the formation of infectious prions, these mutants have provided important insights into possible pathogenic mechanisms. These studies suggest that prion neurotoxicity may involve subversion of a cytoprotective activity of PrP(C) via altered signaling events at the plasma membrane.
Collapse
Affiliation(s)
- Isaac H Solomon
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
44
|
Kim Y, Kim JB, Sohn H, Lee C. A national survey on the allelic, genotypic, and haplotypic distribution of PRNP insertion and deletion polymorphisms in Korean cattle. J Genet 2009; 88:99-103. [PMID: 19417551 DOI: 10.1007/s12041-009-0014-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Younyoung Kim
- Department of Bioinformatics and Life Science, Soongsil University, Seoul 156-743, South Korea
| | | | | | | |
Collapse
|
45
|
Jeffrey M, Goodsir C, McGovern G, Barmada SJ, Medrano AZ, Harris DA. Prion protein with an insertional mutation accumulates on axonal and dendritic plasmalemma and is associated with distinctive ultrastructural changes. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1208-17. [PMID: 19700753 PMCID: PMC2731139 DOI: 10.2353/ajpath.2009.090125] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/19/2009] [Indexed: 11/20/2022]
Abstract
Prion diseases are fatal neurological diseases characterized by central nervous system deposition of abnormal forms of a membrane glycoprotein designated PrP (prion protein). Tg(PG14) transgenic mice express PrP that harbor a nine-octapeptide insertional mutation homologous to one described in a familial prion disease of humans. Tg(PG14) mice spontaneously develop a fatal neurological illness accompanied by massive apoptosis of cerebellar granule neurons and accumulation of an aggregated and weakly protease-resistant form of PrP that is not infectious. Previous light microscopic analyses of these mice left open questions regarding the subcellular distribution of the mutant protein and the nature of the neuropathological lesions produced. To address these questions, we undertook an immunogold electron microscopic study of Tg(PG14) mice. We found that mutant PrP is localized primarily on the plasma membrane of dendrites and unmyelinated axons in the hippocampus and cerebellum, with little labeling of either neuronal cell bodies or intracellular organelles. PrP deposits were shown to be associated with degenerative changes in dendritic structure. We also describe for the first time marked pathology in myelinated axons, and alterations in the axon/oligodendrocyte interface. Taken together, our results suggest cellular mechanisms by which mutant PrPs produce pathology. In addition, they highlight distinctions between familial and infectious prion disorders at the ultrastructural level that correlate with differences in cellular trafficking of the disease-associated PrP forms.
Collapse
Affiliation(s)
- Martin Jeffrey
- Veterinary Laboratories Agency, Lasswade Laboratory, Pentlands Science Park, Bush Loan, Penicuik, Midlothian, Scotland.
| | | | | | | | | | | |
Collapse
|
46
|
Mehrpour M, Codogno P. Prion protein: From physiology to cancer biology. Cancer Lett 2009; 290:1-23. [PMID: 19674833 DOI: 10.1016/j.canlet.2009.07.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Revised: 07/10/2009] [Accepted: 07/13/2009] [Indexed: 12/26/2022]
Abstract
Prion protein (PrPc) was originally viewed solely as being involved in prion disease, but now several intriguing lines of evidence have emerged indicating that it plays a fundamental role not only in the nervous system, but also throughout the human body. PrPc is expressed most abundantly in the brain, but has also been detected in other non-neuronal tissues as diverse as lymphoid cells, lung, heart, kidney, gastrointestinal tract, muscle, and mammary glands. Recent data indicate that PrPc may be implicated in biology of glioblastoma, breast cancer, prostate and gastric cancer. Over expression of PrPc is correlated to the acquisition by tumor cells of a phenotype for resistance to cell death induced by TNF alpha and TRAIL or antitumor drugs such as paclitaxel and anthracyclines. PrPc may promote tumorigenesis, proliferation and G1/S transition in gastric cancer cells. This review revisits the physiological functions of PrPc, and its possible implications for cancer biology.
Collapse
|
47
|
Li XL, Dong CF, Wang GR, Zhou RM, Shi Q, Tian C, Gao C, Mei GY, Chen C, Xu K, Han J, Dong XP. Manganese-induced changes of the biochemical characteristics of the recombinant wild-type and mutant PrPs. Med Microbiol Immunol 2009; 198:239-45. [PMID: 19633867 DOI: 10.1007/s00430-009-0120-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2009] [Indexed: 11/26/2022]
Abstract
Manganese may play some roles in the pathogenesis of prion diseases. In this study, recombinant human wild-type (WT) PrP and PrP mutants with deleted or inserted octarepeats were exposed to manganese, and their biochemical and biophysical characteristics were evaluated by proteinase K (PK) digestion, sedimentation experiments, transmission electron microscopy and circular dichroism. It demonstrated that incubation of manganese remarkably increased PK-resistances, protein aggregations and beta-sheet contents of the PrPs. Moreover, the PrP mutants of inserted or deleted octarepeats were much vulnerable to the influence of manganese, which showed obviously more aggregation and higher beta-sheet content than that of WT-PrP. It highlights that the effect of manganese on the PrP seems to lie on the incorrectness of the octarepeats numbers. The association of the octarepeats number of PrP with manganese may further provide insight into the unresolved biological function of PrP in the neurons.
Collapse
Affiliation(s)
- Xiao-Li Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Stevens DJ, Walter ED, Rodríguez A, Draper D, Davies P, Brown DR, Millhauser GL. Early onset prion disease from octarepeat expansion correlates with copper binding properties. PLoS Pathog 2009; 5:e1000390. [PMID: 19381258 PMCID: PMC2663819 DOI: 10.1371/journal.ppat.1000390] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Accepted: 03/20/2009] [Indexed: 11/18/2022] Open
Abstract
Insertional mutations leading to expansion of the octarepeat domain of the prion protein (PrP) are directly linked to prion disease. While normal PrP has four PHGGGWGQ octapeptide segments in its flexible N-terminal domain, expanded forms may have up to nine additional octapeptide inserts. The type of prion disease segregates with the degree of expansion. With up to four extra octarepeats, the average onset age is above 60 years, whereas five to nine extra octarepeats results in an average onset age between 30 and 40 years, a difference of almost three decades. In wild-type PrP, the octarepeat domain takes up copper (Cu2+) and is considered essential for in vivo function. Work from our lab demonstrates that the copper coordination mode depends on the precise ratio of Cu2+ to protein. At low Cu2+ levels, coordination involves histidine side chains from adjacent octarepeats, whereas at high levels each repeat takes up a single copper ion through interactions with the histidine side chain and neighboring backbone amides. Here we use both octarepeat constructs and recombinant PrP to examine how copper coordination modes are influenced by octarepeat expansion. We find that there is little change in affinity or coordination mode populations for octarepeat domains with up to seven segments (three inserts). However, domains with eight or nine total repeats (four or five inserts) become energetically arrested in the multi-histidine coordination mode, as dictated by higher copper uptake capacity and also by increased binding affinity. We next pooled all published cases of human prion disease resulting from octarepeat expansion and find remarkable agreement between the sudden length-dependent change in copper coordination and onset age. Together, these findings suggest that either loss of PrP copper-dependent function or loss of copper-mediated protection against PrP polymerization makes a significant contribution to early onset prion disease. Prion diseases are neurodegenerative disorders involving the prion protein, a normal component of the central nervous system. An unusual class of inherited mutations giving rise to prion disease involves elongation of the so-called octarepeat domain, near the protein's N-terminus. Research from our lab and others shows that this domain binds the micronutrient copper, an essential element for proper neurological function. We investigated how octarepeat elongation influences copper binding by examining both the molecular features and the binding equilibrium. We find that elongation beyond a specific threshold, which confers profound early onset disease, gives rise to concomitant changes in copper uptake. The remarkable agreement between onset age and altered copper binding points to loss of copper protein function as significant in prion neurodegeneration.
Collapse
Affiliation(s)
- Daniel J. Stevens
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Eric D. Walter
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Abel Rodríguez
- Department of Applied Mathematics and Statistics, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - David Draper
- Department of Applied Mathematics and Statistics, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Paul Davies
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - David R. Brown
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Glenn L. Millhauser
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California, United States of America
- * E-mail:
| |
Collapse
|
49
|
A patient with Creutzfeldt-Jakob disease with an insertion of 7 octa-repeats in the PRNP gene: molecular characteristics and clinical features. Am J Med Sci 2009; 336:519-23. [PMID: 19092329 DOI: 10.1097/maj.0b013e3181643e50] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We evaluated the features of neuropathology, abnormal prion protein (PrP) molecules, and clinical data of a Chinese woman diagnosed with familiar Creutzfeldt-Jakob disease (CJD), having 7 octa-repeats inserted with codon 129 methionine homozygote in the PRNP gene. METHODS Neuropathologic characteristics of the brain were analyzed by hemotoxylin-eosin stain and electronic microscopy. The presence of abnormal PrP in brains was detected by proteinase K and PrP molecules were evaluated by deglycosylation assay. RESULTS Spongiform degeneration, with diffuse neuronal loss and mild astrocytic gliosis, as well as with profound degeneration of neurons and astrocytes was observed. Proteinase K-resistant PrP was deposited widely in various regions of the brain. Calculation of the glycosylation ratios of proteinase K-resistant PrP molecules identified that the monoglycosyl isomer was predominant. PrP deglycosylation tests allowed for the identification of a predominant 19-kDa PrP signal that represents a partially proteolytic C-terminal segment, a 27-kDa band that represents the full-length wild-type PrP molecule, and a 30-kDa band that probably corresponds to the full-length mutant PrP molecule. CONCLUSION : Sporadic CJD-like neuropathologic changes and deposits of proteinase K-resistant PrP have been identified in this familiar CJD case with a 168 base pair nucleotide insertion. The clinical features differ from previously reported cases that had 7 octa-repeat insertion, but bear similarities to sporadic CJD.
Collapse
|
50
|
De novo generation of a transmissible spongiform encephalopathy by mouse transgenesis. Proc Natl Acad Sci U S A 2008; 106:304-9. [PMID: 19073920 DOI: 10.1073/pnas.0810680105] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Most transmissible spongiform encephalopathies arise either spontaneously or by infection. Mutations of PRNP, which encodes the prion protein, PrP, segregate with phenotypically similar diseases. Here we report that moderate overexpression in transgenic mice of mPrP(170N,174T), a mouse PrP with two point mutations that subtly affect the structure of its globular domain, causes a fully penetrant lethal spongiform encephalopathy with cerebral PrP plaques. This genetic disease was reproduced with 100% attack rate by intracerebral inoculation of brain homogenate to tga20 mice overexpressing WT PrP, and from the latter to WT mice, but not to PrP-deficient mice. Upon successive transmissions, the incubation periods decreased and PrP became more protease-resistant, indicating the presence of a strain barrier that was gradually overcome by repeated passaging. This shows that expression of a subtly altered prion protein, with known 3D structure, efficiently generates a prion disease.
Collapse
|