1
|
Napper S, Schatzl HM. Oral vaccination as a potential strategy to manage chronic wasting disease in wild cervid populations. Front Immunol 2023; 14:1156451. [PMID: 37122761 PMCID: PMC10140515 DOI: 10.3389/fimmu.2023.1156451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Prion diseases are a novel class of infectious disease based in the misfolding of the cellular prion protein (PrPC) into a pathological, self-propagating isoform (PrPSc). These fatal, untreatable neurodegenerative disorders affect a variety of species causing scrapie in sheep and goats, bovine spongiform encephalopathy (BSE) in cattle, chronic wasting disease (CWD) in cervids, and Creutzfeldt-Jacob disease (CJD) in humans. Of the animal prion diseases, CWD is currently regarded as the most significant threat due its ongoing geographical spread, environmental persistence, uptake into plants, unpredictable evolution, and emerging evidence of zoonotic potential. The extensive efforts to manage CWD have been largely ineffective, highlighting the need for new disease management tools, including vaccines. Development of an effective CWD vaccine is challenged by the unique biology of these diseases, including the necessity, and associated dangers, of overcoming immune tolerance, as well the logistical challenges of vaccinating wild animals. Despite these obstacles, there has been encouraging progress towards the identification of safe, protective antigens as well as effective strategies of formulation and delivery that would enable oral delivery to wild cervids. In this review we highlight recent strategies for antigen selection and optimization, as well as considerations of various platforms for oral delivery, that will enable researchers to accelerate the rate at which candidate CWD vaccines are developed and evaluated.
Collapse
Affiliation(s)
- Scott Napper
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Hermann M. Schatzl
- Calgary Prion Research Unit, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
2
|
Mathiason CK. Large animal models for chronic wasting disease. Cell Tissue Res 2023; 392:21-31. [PMID: 35113219 PMCID: PMC8811588 DOI: 10.1007/s00441-022-03590-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/19/2022] [Indexed: 11/30/2022]
Abstract
Chronic wasting disease (CWD) is a fatal neurodegenerative prion disease of cervid species including deer, elk, moose and reindeer. The disease has shown both geographic and species expansion since its discovery in the late 1960's and is now recognized in captive and free-ranging cervid populations in North America, Asia and Europe. The facile transmission of CWD is unique among prion diseases and has resulted in growing concern for cervid populations and human public health. The development of native cervid host models with longitudinal monitoring has revealed new insights about CWD pathogenesis and transmission dynamics. More than 20 years of experimental studies conducted in these models, using biologically relevant routes of infection, have led to better understanding of many aspect of CWD infections. This review addresses some of these insights, including: (i) the temporal intra-host trafficking of CWD prions in tissues and bodily fluids, (ii) the presence of infectivity shed in bodily excretions that may help explain the facile transmission of CWD, (iii) mother-to-offspring CWD transmission, (iv) the influence of some Prnp polymorphisms on CWD susceptibility, and (vi) continued development of vaccine strategies to mitigate CWD.
Collapse
Affiliation(s)
- C K Mathiason
- College of Veterinary Medicine and Biomedical Sciences, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States, 80523.
| |
Collapse
|
3
|
Triller G, Garyfallos DA, Papavasiliou FN, Sklaviadis T, Stavropoulos P, Xanthopoulos K. Immunization with Genetically Modified Trypanosomes Provides Protection against Transmissible Spongiform Encephalopathies. Int J Mol Sci 2022; 23:ijms231810629. [PMID: 36142526 PMCID: PMC9503410 DOI: 10.3390/ijms231810629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Transmissible spongiform encephalopathies are incurable neurodegenerative diseases, associated with the conversion of the physiological prion protein to its disease-associated counterpart. Even though immunization against transmissible spongiform encephalopathies has shown great potential, immune tolerance effects impede the use of active immunization protocols for successful prophylaxis. In this study, we evaluate the use of trypanosomes as biological platforms for the presentation of a prion antigenic peptide to the host immune system. Using the engineered trypanosomes in an immunization protocol without the use of adjuvants led to the development of a humoral immune response against the prion protein in wild type mice, without the appearance of adverse reactions. The immune reaction elicited with this protocol displayed in vitro therapeutic potential and was further evaluated in a bioassay where immunized mice were partially protected in a representative murine model of prion diseases. Further studies are underway to better characterize the immune reaction and optimize the immunization protocol.
Collapse
Affiliation(s)
- Gianna Triller
- Laboratory of Lymphocyte Biology, The Rockefeller University, New York, NY 10065, USA
| | - Dimitrios A. Garyfallos
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - F. Nina Papavasiliou
- Division of Immune Diversity, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| | - Theodoros Sklaviadis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Pete Stavropoulos
- Laboratory of Lymphocyte Biology, The Rockefeller University, New York, NY 10065, USA
- Correspondence: (P.S.); (K.X.); Tel.: +30-2310-997-654 (Κ.Χ.)
| | - Konstantinos Xanthopoulos
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, 57001 Thermi, Greece
- Correspondence: (P.S.); (K.X.); Tel.: +30-2310-997-654 (Κ.Χ.)
| |
Collapse
|
4
|
Mohammadi B, Song F, Matamoros-Angles A, Shafiq M, Damme M, Puig B, Glatzel M, Altmeppen HC. Anchorless risk or released benefit? An updated view on the ADAM10-mediated shedding of the prion protein. Cell Tissue Res 2022; 392:215-234. [PMID: 35084572 PMCID: PMC10113312 DOI: 10.1007/s00441-022-03582-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/12/2022] [Indexed: 11/24/2022]
Abstract
The prion protein (PrP) is a broadly expressed glycoprotein linked with a multitude of (suggested) biological and pathological implications. Some of these roles seem to be due to constitutively generated proteolytic fragments of the protein. Among them is a soluble PrP form, which is released from the surface of neurons and other cell types by action of the metalloprotease ADAM10 in a process termed 'shedding'. The latter aspect is the focus of this review, which aims to provide a comprehensive overview on (i) the relevance of proteolytic processing in regulating cellular PrP functions, (ii) currently described involvement of shed PrP in neurodegenerative diseases (including prion diseases and Alzheimer's disease), (iii) shed PrP's expected roles in intercellular communication in many more (patho)physiological conditions (such as stroke, cancer or immune responses), (iv) and the need for improved research tools in respective (future) studies. Deeper mechanistic insight into roles played by PrP shedding and its resulting fragment may pave the way for improved diagnostics and future therapeutic approaches in diseases of the brain and beyond.
Collapse
Affiliation(s)
- Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- Working Group for Interdisciplinary Neurobiology and Immunology (INI Research), Hamburg, Germany
| | - Feizhi Song
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Andreu Matamoros-Angles
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Damme
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | |
Collapse
|
5
|
Linsenmeier L, Mohammadi B, Shafiq M, Frontzek K, Bär J, Shrivastava AN, Damme M, Song F, Schwarz A, Da Vela S, Massignan T, Jung S, Correia A, Schmitz M, Puig B, Hornemann S, Zerr I, Tatzelt J, Biasini E, Saftig P, Schweizer M, Svergun D, Amin L, Mazzola F, Varani L, Thapa S, Gilch S, Schätzl H, Harris DA, Triller A, Mikhaylova M, Aguzzi A, Altmeppen HC, Glatzel M. Ligands binding to the prion protein induce its proteolytic release with therapeutic potential in neurodegenerative proteinopathies. SCIENCE ADVANCES 2021; 7:eabj1826. [PMID: 34818048 PMCID: PMC8612689 DOI: 10.1126/sciadv.abj1826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/20/2021] [Indexed: 05/07/2023]
Abstract
The prion protein (PrPC) is a central player in neurodegenerative diseases, such as prion diseases or Alzheimer’s disease. In contrast to disease-promoting cell surface PrPC, extracellular fragments act neuroprotective by blocking neurotoxic disease-associated protein conformers. Fittingly, PrPC release by the metalloprotease ADAM10 represents a protective mechanism. We used biochemical, cell biological, morphological, and structural methods to investigate mechanisms stimulating this proteolytic shedding. Shed PrP negatively correlates with prion conversion and is markedly redistributed in murine brain in the presence of prion deposits or amyloid plaques, indicating a sequestrating activity. PrP-directed ligands cause structural changes in PrPC and increased shedding in cells and organotypic brain slice cultures. As an exception, some PrP-directed antibodies targeting repetitive epitopes do not cause shedding but surface clustering, endocytosis, and degradation of PrPC. Both mechanisms may contribute to beneficial actions described for PrP-directed ligands and pave the way for new therapeutic strategies against currently incurable neurodegenerative diseases.
Collapse
Affiliation(s)
- Luise Linsenmeier
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Karl Frontzek
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Julia Bär
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Molecular Neurobiology Hamburg (ZMNH), UKE, Hamburg, Germany
| | - Amulya N. Shrivastava
- École Normale Supérieure, Institut de Biologie de l’ENS (IBENS), INSERM, CNRS, PSL Research University, Paris, France
| | - Markus Damme
- Institute of Biochemistry, Christian Albrechts University, Kiel, Germany
| | - Feizhi Song
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Alexander Schwarz
- Institute of Nanostructure and Solid State Physics, Universität Hamburg, Hamburg, Germany
| | - Stefano Da Vela
- European Molecular Biology Laboratory (EMBL), Hamburg, Germany
| | - Tania Massignan
- Dulbecco Telethon Laboratory of Prions and Amyloids, CIBIO, University of Trento, Trento, Italy
| | - Sebastian Jung
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Angela Correia
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Matthias Schmitz
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation, UKE, Hamburg, Germany
| | - Simone Hornemann
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jörg Tatzelt
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Cluster of Excellence RESOLV, Bochum, Germany
| | - Emiliano Biasini
- Dulbecco Telethon Laboratory of Prions and Amyloids, CIBIO, University of Trento, Trento, Italy
| | - Paul Saftig
- Institute of Biochemistry, Christian Albrechts University, Kiel, Germany
| | | | - Dmitri Svergun
- European Molecular Biology Laboratory (EMBL), Hamburg, Germany
| | - Ladan Amin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Federica Mazzola
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Luca Varani
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Simrika Thapa
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, Canada
| | - Sabine Gilch
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, Canada
| | - Hermann Schätzl
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, Canada
| | - David A. Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Antoine Triller
- École Normale Supérieure, Institut de Biologie de l’ENS (IBENS), INSERM, CNRS, PSL Research University, Paris, France
| | - Marina Mikhaylova
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Molecular Neurobiology Hamburg (ZMNH), UKE, Hamburg, Germany
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Hermann C. Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
6
|
Eiden M, Gedvilaite A, Leidel F, Ulrich RG, Groschup MH. Vaccination with Prion Peptide-Displaying Polyomavirus-Like Particles Prolongs Incubation Time in Scrapie-Infected Mice. Viruses 2021; 13:v13050811. [PMID: 33946367 PMCID: PMC8147134 DOI: 10.3390/v13050811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/22/2022] Open
Abstract
Prion diseases like scrapie in sheep, bovine spongiform encephalopathy (BSE) in cattle or Creutzfeldt–Jakob disease (CJD) in humans are fatal neurodegenerative diseases characterized by the conformational conversion of the normal, mainly α-helical cellular prion protein (PrPC) into the abnormal β-sheet rich infectious isoform PrPSc. Various therapeutic or prophylactic approaches have been conducted, but no approved therapeutic treatment is available so far. Immunisation against prions is hampered by the self-tolerance to PrPC in mammalian species. One strategy to avoid this tolerance is presenting PrP variants in virus-like particles (VLPs). Therefore, we vaccinated C57/BL6 mice with nine prion peptide variants presented by hamster polyomavirus capsid protein VP1/VP2-derived VLPs. Mice were subsequently challenged intraperitoneally with the murine RML prion strain. Importantly, one group exhibited significantly increased mean survival time of 240 days post-inoculation compared with 202 days of the control group. These data show that immunisation with VLPs presenting PrP peptides may represent a promising strategy for an effective vaccination against transmissible spongiform encephalitis agents.
Collapse
Affiliation(s)
- Martin Eiden
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (F.L.); (R.G.U.); (M.H.G.)
- Correspondence:
| | - Alma Gedvilaite
- Life Sciences Center, Institute of Biotechnology, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania;
| | - Fabienne Leidel
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (F.L.); (R.G.U.); (M.H.G.)
- Task Force Animal Diseases, Darmstadt Regional Administrative Council, Luisenplatz 2, 64283 Darmstadt, Germany
| | - Rainer G. Ulrich
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (F.L.); (R.G.U.); (M.H.G.)
| | - Martin H. Groschup
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (F.L.); (R.G.U.); (M.H.G.)
| |
Collapse
|
7
|
Internalization of α-synuclein oligomers into SH-SY5Y cells. Biophys J 2021; 120:877-885. [PMID: 33515601 DOI: 10.1016/j.bpj.2020.12.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/09/2020] [Accepted: 12/30/2020] [Indexed: 11/20/2022] Open
Abstract
Aggregates of misfolded α-synuclein are a distinctive feature of Parkinson's disease. Small oligomers of α-synuclein are thought to be an important neurotoxic agent, and α-synuclein aggregates exhibit prion-like behavior, propagating misfolding between cells. α-Synuclein is internalized by both passive diffusion and active uptake mechanisms, but how uptake varies with the size of the oligomer is less clear. We explored how α-synuclein internalization into live SH-SY5Y cells varied with oligomer size by comparing the uptake of fluorescently labeled monomers to that of engineered tandem dimers and tetramers. We found that these α-synuclein constructs were internalized primarily through endocytosis. Oligomer size had little effect on their internalization pathway, whether they were added individually or together. Measurements of co-localization of the α-synuclein constructs with fluorescent markers for early endosomes and lysosomes showed that most of the α-synuclein entered endocytic compartments, in which they were probably degraded. Treatment of the cells with the Pitstop inhibitor suggested that most of the oligomers were internalized by the clathrin-mediated pathway.
Collapse
|
8
|
Ma Y, Ma J. Immunotherapy against Prion Disease. Pathogens 2020; 9:E216. [PMID: 32183309 PMCID: PMC7157205 DOI: 10.3390/pathogens9030216] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/12/2020] [Accepted: 03/12/2020] [Indexed: 11/17/2022] Open
Abstract
The term "prion disease" encompasses a group of neurodegenerative diseases affecting both humans and animals. Currently, there is no effective therapy and all forms of prion disease are invariably fatal. Because of (a) the outbreak of bovine spongiform encephalopathy in cattle and variant Creutzfeldt-Jakob disease in humans; (b) the heated debate about the prion hypothesis; and (c) the availability of a natural prion disease in rodents, the understanding of the pathogenic process in prion disease is much more advanced compared to that of other neurodegenerative disorders, which inspired many attempts to develop therapeutic strategies against these fatal diseases. In this review, we focus on immunotherapy against prion disease. We explain our rationale for immunotherapy as a plausible therapeutic choice, review previous trials using either active or passive immunization, and discuss potential strategies for overcoming the hurdles in developing a successful immunotherapy. We propose that immunotherapy is a plausible and practical therapeutic strategy and advocate more studies in this area to develop effective measures to control and treat these devastating disorders.
Collapse
Affiliation(s)
| | - Jiyan Ma
- Center for Neurodegenerative Science, Van Andel Institute, 333 Bostwick Avenue N.E., Grand Rapids, MI 49503, USA;
| |
Collapse
|
9
|
Sengupta I, Udgaonkar J. Monitoring site-specific conformational changes in real-time reveals a misfolding mechanism of the prion protein. eLife 2019; 8:44698. [PMID: 31232689 PMCID: PMC6590988 DOI: 10.7554/elife.44698] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 06/11/2019] [Indexed: 11/25/2022] Open
Abstract
During pathological aggregation, proteins undergo remarkable conformational re-arrangements to anomalously assemble into a heterogeneous collection of misfolded multimers, ranging from soluble oligomers to insoluble amyloid fibrils. Inspired by fluorescence resonance energy transfer (FRET) measurements of protein folding, an experimental strategy to study site-specific misfolding kinetics during aggregation, by effectively suppressing contributions from inter-molecular FRET, is described. Specifically, the kinetics of conformational changes across different secondary and tertiary structural segments of the mouse prion protein (moPrP) were monitored independently, after the monomeric units transformed into large oligomers OL, which subsequently disaggregated reversibly into small oligomers OS at pH 4. The sequence segments spanning helices α2 and α3 underwent a compaction during the formation of OL and elongation into β-sheets during the formation of OS. The β1-α1-β2 and α2-α3 subdomains were separated, and the helix α1 was unfolded to varying extents in both OL and OS.
Collapse
Affiliation(s)
- Ishita Sengupta
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India
| | - Jayant Udgaonkar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India
| |
Collapse
|
10
|
Shen N, Song G, Yang H, Lin X, Brown B, Hong Y, Cai J, Cao C. Identifying the Pathological Domain of Alpha- Synuclein as a Therapeutic for Parkinson's Disease. Int J Mol Sci 2019; 20:E2338. [PMID: 31083520 PMCID: PMC6539124 DOI: 10.3390/ijms20092338] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/02/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022] Open
Abstract
Alpha-synuclein is considered the major pathological protein associated with Parkinson's disease, but there is still no effective immunotherapy which targets alpha-synuclein. In order to create a safer and more effective therapy against PD, we are targeting an epitope of alpha-synuclein rather than full-length alpha-synuclein. We have selected several antigenic domains (B-cell epitope) through antigenicity prediction, and also made several recombinant protein fragments from alpha-synuclein upon antigenicity prediction in an E. coli system. We then tested the function of each of the peptides and recombinant fragments in aggregation, their toxicity and antigenicity. We have discovered that the full-length recombinant (aa1-140) can aggregate into oligomers or even fibrils, and fragment aa15-65 can promote the aggregation of aa1-140. It is worth noting that it not only promotes whole protein aggregation, but also self-aggregates as seen by western blotting and silver staining assays. We have tested all candidates on primary neurons for their toxicity and discovered that aa15-65 is the most toxic domain compared to all other fragments. The antibody targeting this domain also showed both anti-aggregation activity and some therapeutic effect. Therefore, we believe that we have identified the most potent therapeutic domain of alpha synuclein as a therapeutic target.
Collapse
Affiliation(s)
- Ning Shen
- Department of Chemistry, University of South Florida, Tampa, FL 33612, USA.
| | - Ge Song
- Department of Surgery of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Haiqiang Yang
- Department of Chemistry, University of South Florida, Tampa, FL 33612, USA.
| | - Xiaoyang Lin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA.
| | - Breanna Brown
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA.
| | - Yuzhu Hong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA.
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL 33612, USA.
| | - Chuanhai Cao
- Department of Chemistry, University of South Florida, Tampa, FL 33612, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA.
- Department of Neurology, College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
11
|
Forloni G, Roiter I, Tagliavini F. Clinical trials of prion disease therapeutics. Curr Opin Pharmacol 2019; 44:53-60. [DOI: 10.1016/j.coph.2019.04.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 12/31/2022]
|
12
|
Abdelaziz DH, Thapa S, Brandon J, Maybee J, Vankuppeveld L, McCorkell R, Schätzl HM. Recombinant prion protein vaccination of transgenic elk PrP mice and reindeer overcomes self-tolerance and protects mice against chronic wasting disease. J Biol Chem 2018; 293:19812-19822. [PMID: 30397182 PMCID: PMC6314114 DOI: 10.1074/jbc.ra118.004810] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/29/2018] [Indexed: 12/17/2022] Open
Abstract
Chronic wasting disease (CWD) is a fatal neurodegenerative disease that affects cervids in North America and now Europe. No effective measures are available to control CWD. We hypothesized that active vaccination with homologous and aggregation-prone recombinant prion protein (PrP) could overcome self-tolerance and induce autoantibody production against the cellular isoform of PrP (PrPC), which would be protective against CWD infection from peripheral routes. Five groups of transgenic mice expressing elk PrP (TgElk) were vaccinated with either the adjuvant CpG alone or one of four recombinant PrP immunogens: deer dimer (Ddi); deer monomer (Dmo); mouse dimer (Mdi); and mouse monomer (Mmo). Mice were then challenged intraperitoneally with elk CWD prions. All vaccinated mice developed ELISA-detectable antibody titers against PrP. Importantly, all four vaccinated groups survived longer than the control group, with the Mmo-immunized group exhibiting 60% prolongation of mean survival time compared with the control group (183 versus 114 days post-inoculation). We tested for prion infection in brain and spleen of all clinically sick mice. Notably, the attack rate was 100% as revealed by positive CWD signals in all tested tissues when assessed with Western blotting, real-time quaking-induced conversion, and immunohistochemistry. Our pilot study in reindeer indicated appreciable humoral immune responses to Mdi and Ddi immunogens, and the post-immune sera from the Ddi-vaccinated reindeer mitigated CWD propagation in a cell culture model (CWD-RK13). Taken together, our study provides very promising vaccine candidates against CWD, but further studies in cervids are required to investigate vaccine efficacy in the natural CWD hosts.
Collapse
Affiliation(s)
- Dalia H Abdelaziz
- From the Department of Comparative Biology and Experimental Medicine and.,the Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt.,the Calgary Prion Research Unit, University of Calgary, Calgary, Alberta T2N 4Z6, Canada and
| | - Simrika Thapa
- From the Department of Comparative Biology and Experimental Medicine and.,the Calgary Prion Research Unit, University of Calgary, Calgary, Alberta T2N 4Z6, Canada and
| | - Jenna Brandon
- From the Department of Comparative Biology and Experimental Medicine and
| | - Justine Maybee
- From the Department of Comparative Biology and Experimental Medicine and
| | | | - Robert McCorkell
- From the Department of Comparative Biology and Experimental Medicine and
| | - Hermann M Schätzl
- From the Department of Comparative Biology and Experimental Medicine and .,the Calgary Prion Research Unit, University of Calgary, Calgary, Alberta T2N 4Z6, Canada and
| |
Collapse
|
13
|
Interaction of Peptide Aptamers with Prion Protein Central Domain Promotes α-Cleavage of PrP C. Mol Neurobiol 2018; 55:7758-7774. [PMID: 29460268 PMCID: PMC6132731 DOI: 10.1007/s12035-018-0944-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 01/31/2018] [Indexed: 11/03/2022]
Abstract
Prion diseases are infectious and fatal neurodegenerative diseases affecting humans and animals. Transmission is possible within and between species with zoonotic potential. Currently, no prophylaxis or treatment exists. Prions are composed of the misfolded isoform PrPSc of the cellular prion protein PrPC. Expression of PrPC is a prerequisite for prion infection, and conformational conversion of PrPC is induced upon its direct interaction with PrPSc. Inhibition of this interaction can abrogate prion propagation, and we have previously established peptide aptamers (PAs) binding to PrPC as new anti-prion compounds. Here, we mapped the interaction site of PA8 in PrP and modeled the complex in silico to design targeted mutations in PA8 which presumably enhance binding properties. Using these PA8 variants, we could improve PA-mediated inhibition of PrPSc replication and de novo infection of neuronal cells. Furthermore, we demonstrate that binding of PA8 and its variants increases PrPC α-cleavage and interferes with its internalization. This gives rise to high levels of the membrane-anchored PrP-C1 fragment, a transdominant negative inhibitor of prion replication. PA8 and its variants interact with PrPC at its central and most highly conserved domain, a region which is crucial for prion conversion and facilitates toxic signaling of Aβ oligomers characteristic for Alzheimer's disease. Our strategy allows for the first time to induce α-cleavage, which occurs within this central domain, independent of targeting the responsible protease. Therefore, interaction of PAs with PrPC and enhancement of α-cleavage represent mechanisms that can be beneficial for the treatment of prion and other neurodegenerative diseases.
Collapse
|
14
|
Abstract
Currently all prion diseases are without effective treatment and are universally fatal. It is increasingly being recognized that the pathogenesis of many neurodegenerative diseases, such as Alzheimer disease (AD), includes "prion-like" properties. Hence, any effective therapeutic intervention for prion disease could have significant implications for other neurodegenerative diseases. Conversely, therapies that are effective in AD might also be therapeutically beneficial for prion disease. AD-like prion disease has no effective therapy. However, various vaccine and immunomodulatory approaches have shown great success in animal models of AD, with numerous ongoing clinical trials of these potential immunotherapies. More limited evidence suggests that immunotherapies may be effective in prion models and in naturally occurring prion disease. In particular, experimental data suggest that mucosal vaccination against prions can be effective for protection against orally acquired prion infection. Many prion diseases, including natural sheep scrapie, bovine spongiform encephalopathy, chronic wasting disease, and variant Creutzfeldt-Jakob disease, are thought to be acquired peripherally, mainly by oral exposure. Mucosal vaccination would be most applicable to this form of transmission. In this chapter we review various immunologically based strategies which are under development for prion infection.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Center for Cognitive Neurology, New York University School of Medicine, New York, NY, United States; Department of Neurology, New York University School of Medicine, New York, NY, United States; Department of Pathology, New York University School of Medicine, New York, NY, United States; Department of Psychiatry, New York University School of Medicine, New York, NY, United States.
| | - Fernando Goñi
- Center for Cognitive Neurology, New York University School of Medicine, New York, NY, United States; Department of Neurology, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
15
|
Charco JM, Eraña H, Venegas V, García-Martínez S, López-Moreno R, González-Miranda E, Pérez-Castro MÁ, Castilla J. Recombinant PrP and Its Contribution to Research on Transmissible Spongiform Encephalopathies. Pathogens 2017; 6:E67. [PMID: 29240682 PMCID: PMC5750591 DOI: 10.3390/pathogens6040067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/08/2017] [Accepted: 12/12/2017] [Indexed: 01/13/2023] Open
Abstract
The misfolding of the cellular prion protein (PrPC) into the disease-associated isoform (PrPSc) and its accumulation as amyloid fibrils in the central nervous system is one of the central events in transmissible spongiform encephalopathies (TSEs). Due to the proteinaceous nature of the causal agent the molecular mechanisms of misfolding, interspecies transmission, neurotoxicity and strain phenomenon remain mostly ill-defined or unknown. Significant advances were made using in vivo and in cellula models, but the limitations of these, primarily due to their inherent complexity and the small amounts of PrPSc that can be obtained, gave rise to the necessity of new model systems. The production of recombinant PrP using E. coli and subsequent induction of misfolding to the aberrant isoform using different techniques paved the way for the development of cell-free systems that complement the previous models. The generation of the first infectious recombinant prion proteins with identical properties of brain-derived PrPSc increased the value of cell-free systems for research on TSEs. The versatility and ease of implementation of these models have made them invaluable for the study of the molecular mechanisms of prion formation and propagation, and have enabled improvements in diagnosis, high-throughput screening of putative anti-prion compounds and the design of novel therapeutic strategies. Here, we provide an overview of the resultant advances in the prion field due to the development of recombinant PrP and its use in cell-free systems.
Collapse
Affiliation(s)
- Jorge M. Charco
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; (J.M.C.); (H.E.); (V.V.); (S.G.-M.); (R.L.-M.); (E.G.-M.); (M.Á.P.-C.)
| | - Hasier Eraña
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; (J.M.C.); (H.E.); (V.V.); (S.G.-M.); (R.L.-M.); (E.G.-M.); (M.Á.P.-C.)
| | - Vanessa Venegas
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; (J.M.C.); (H.E.); (V.V.); (S.G.-M.); (R.L.-M.); (E.G.-M.); (M.Á.P.-C.)
| | - Sandra García-Martínez
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; (J.M.C.); (H.E.); (V.V.); (S.G.-M.); (R.L.-M.); (E.G.-M.); (M.Á.P.-C.)
| | - Rafael López-Moreno
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; (J.M.C.); (H.E.); (V.V.); (S.G.-M.); (R.L.-M.); (E.G.-M.); (M.Á.P.-C.)
| | - Ezequiel González-Miranda
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; (J.M.C.); (H.E.); (V.V.); (S.G.-M.); (R.L.-M.); (E.G.-M.); (M.Á.P.-C.)
| | - Miguel Ángel Pérez-Castro
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; (J.M.C.); (H.E.); (V.V.); (S.G.-M.); (R.L.-M.); (E.G.-M.); (M.Á.P.-C.)
| | - Joaquín Castilla
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; (J.M.C.); (H.E.); (V.V.); (S.G.-M.); (R.L.-M.); (E.G.-M.); (M.Á.P.-C.)
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| |
Collapse
|
16
|
Abstract
Protein sequences are evolved to encode generally one folded structure, out of a nearly infinite array of possible folds. Underlying this code is a funneled free energy landscape that guides folding to the native conformation. Protein misfolding and aggregation are also a manifestation of free-energy landscapes. The detailed mechanisms of these processes are poorly understood, but often involve rare, transient species and a variety of different pathways. The inherent complexity of misfolding has hampered efforts to measure aggregation pathways and the underlying energy landscape, especially using traditional methods where ensemble averaging obscures important rare and transient events. We recently studied the misfolding and aggregation of prion protein by examining 2 monomers tethered in close proximity as a dimer, showing how the steps leading to the formation of a stable aggregated state can be resolved in the single-molecule limit and the underlying energy landscape thereby reconstructed. This approach allows a more quantitative comparison of native folding versus misfolding, including fundamental differences in the dynamics for misfolding. By identifying key steps and interactions leading to misfolding, it should help to identify potential drug targets. Here we describe the importance of characterizing free-energy landscapes for aggregation and the challenges involved in doing so, and we discuss how single-molecule studies can help test proposed structural models for PrP aggregates.
Collapse
Affiliation(s)
- Derek R Dee
- a Department of Physics , University of Alberta , Edmonton , AB , Canada
| | - Michael T Woodside
- a Department of Physics , University of Alberta , Edmonton , AB , Canada;,b National Institute for Nanotechnology, National Research Council , Edmonton , AB , Canada
| |
Collapse
|
17
|
Immunization of cervidized transgenic mice with multimeric deer prion protein induces self-antibodies that antagonize chronic wasting disease infectivity in vitro. Sci Rep 2017; 7:10538. [PMID: 28874781 PMCID: PMC5585258 DOI: 10.1038/s41598-017-11235-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/18/2017] [Indexed: 12/14/2022] Open
Abstract
Chronic wasting disease (CWD) is the most contagious prion disease. It is expanding rapidly in North America, was found recently in Europe, and the potential for transmission to humans cannot be excluded yet. We hypothesized that it is possible to prevent peripheral CWD infection and CWD prion shedding by inducing auto-antibodies against the cellular prion protein (PrPC) by active vaccination. Our objective is to overcome self-tolerance against PrP by using a multimeric recombinant PrP (recPrP) as an immunogen. We expressed in E. coli, purified and refolded four immunogens: cervid and murine recPrP in monomeric and dimeric form. Testing immunogenicity in sera of the vaccinated transgenic mice expressing cervid PrP revealed that all four immunogens effectively overcame self-tolerance against the prion protein as shown by high antibody titers. Confocal microscopy analysis revealed effective binding of post-immune sera to surface-located PrPC in both murine and cervid PrP expressing cultured cells. Remarkably, the post-immune auto-antibodies effectively inhibited CWD-induced prion conversion in RT-QuIC assay when incubated with either PrP substrate or CWD seed. Furthermore, they mitigated prion propagation in CWD-infected cervid-PrP expressing RK13 cells. Together, multimeric recombinant cervid PrP effectively overcomes self-tolerance to PrP and induces auto-antibodies that interfere with CWD conversion in vitro.
Collapse
|
18
|
Abstract
Prion diseases denote a distinct form of infectivity that is based in the misfolding of a self-protein (PrP(C)) into a pathological, infectious conformation (PrP(Sc)). Efforts to develop vaccines for prion diseases have been complicated by the potential dangers that are associated with induction of immune responses against a self-protein. As a consequence, there is considerable appeal for vaccines that specifically target the misfolded prion conformation. Such conformation-specific immunotherapy is made possible through the identification of vaccine targets (epitopes) that are exclusively presented as a consequence of misfolding. An immune response directed against these targets, termed disease-specific epitopes (DSEs), has the potential to spare the function of the native form of the protein while clearing, or neutralizing, the infectious isomer. Although identification of DSEs represents a critical first step in the induction of conformation-specific immune responses, substantial efforts are required to translate these targets into functional vaccines. Due to the poor immunogenicity that is inherent to self-proteins, and that is often associated with short peptides, substantial efforts are required to overcome tolerance-to-self and maximize the resultant immune response following DSE-based immunization. This often includes optimization of target sequences in terms of immunogenicity and development of effective formulation and delivery strategies for the associated peptides. Further, these vaccines must satisfy additional criteria from perspectives of specificity (PrP(C) vs. PrP(Sc)) and safety (antibody-induced template-driven misfolding of PrP(C)). The emphasis of this report is on the steps required to translate DSEs into prion vaccines and subsequent evaluation of the resulting immune responses.
Collapse
|
19
|
Protein misfolding occurs by slow diffusion across multiple barriers in a rough energy landscape. Proc Natl Acad Sci U S A 2015; 112:8308-13. [PMID: 26109573 DOI: 10.1073/pnas.1419197112] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The timescale for the microscopic dynamics of proteins during conformational transitions is set by the intrachain diffusion coefficient, D. Despite the central role of protein misfolding and aggregation in many diseases, it has proven challenging to measure D for these processes because of their heterogeneity. We used single-molecule force spectroscopy to overcome these challenges and determine D for misfolding of the prion protein PrP. Observing directly the misfolding of individual dimers into minimal aggregates, we reconstructed the energy landscape governing nonnative structure formation. Remarkably, rather than displaying multiple pathways, as typically expected for aggregation, PrP dimers were funneled into a thermodynamically stable misfolded state along a single pathway containing several intermediates, one of which blocked native folding. Using Kramers' rate theory, D was found to be 1,000-fold slower for misfolding than for native folding, reflecting local roughening of the misfolding landscape, likely due to increased internal friction. The slow diffusion also led to much longer transit times for barrier crossing, allowing transition paths to be observed directly for the first time to our knowledge. These results open a new window onto the microscopic mechanisms governing protein misfolding.
Collapse
|
20
|
Rouget R, Sharma G, LeBlanc AC. Cyclin-dependent kinase 5 phosphorylation of familial prion protein mutants exacerbates conversion into amyloid structure. J Biol Chem 2015; 290:5759-71. [PMID: 25572400 DOI: 10.1074/jbc.m114.630699] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Familial prion protein (PrP) mutants undergo conversion from soluble and protease-sensitive to insoluble and partially protease-resistant proteins. Cyclin-dependent kinase 5 (Cdk5) phosphorylation of wild type PrP (pPrP) at serine 43 induces a conversion of PrP into aggregates and fibrils. Here, we investigated whether familial PrP mutants are predisposed to Cdk5 phosphorylation and whether phosphorylation of familial PrP mutants increases conversion. PrP mutants representing three major familial PrP diseases and different PrP structural domains were studied. We developed a novel in vitro kinase reaction coupled with Thioflavin T binding to amyloid structure assay to monitor phosphorylation-dependent amyloid conversion. Although non-phosphorylated full-length wild type or PrP mutants did not convert into amyloid, Cdk5 phosphorylation rapidly converted these into Thioflavin T-positive structures following first order kinetics. Dephosphorylation partially reversed conversion. Phosphorylation-dependent conversion of PrP from α-helical structures into β-sheet structures was confirmed by circular dichroism. Relative to wild type pPrP, most PrP mutants showed increased rate constants of conversion. In contrast, non-phosphorylated truncated PrP Y145X (where X represents a stop codon) and Q160X mutants converted spontaneously into Thioflavin T-positive fibrils after a lag phase of over 20 h, indicating nucleation-dependent polymerization. Phosphorylation reduced the lag phase by over 50% and thus accelerated the formation of the nucleating event. Consistently, phosphorylated Y145X and phosphorylated Q160X exacerbated conversion in a homologous seeding reaction, whereas WT pPrP could not seed WT PrP. These results demonstrate an influence of both the N terminus and the C terminus of PrP on conversion. We conclude that post-translational modifications of the flexible N terminus of PrP can cause or exacerbate PrP mutant conversion.
Collapse
Affiliation(s)
- Raphaël Rouget
- From the Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Department of Neurology and Neurosurgery, McGill University, Montréal, Québec H3T 1E2, Canada and
| | - Gyanesh Sharma
- From the Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Department of Neurology and Neurosurgery, McGill University, Montréal, Québec H3T 1E2, Canada and Department of Neurology and Neurosurgery, McGill University, 3775 University Street, Montréal, Québec H3A 2B4, Canada
| | - Andréa C LeBlanc
- From the Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, Department of Neurology and Neurosurgery, McGill University, Montréal, Québec H3T 1E2, Canada and Department of Neurology and Neurosurgery, McGill University, 3775 University Street, Montréal, Québec H3A 2B4, Canada
| |
Collapse
|
21
|
Goñi F, Mathiason CK, Yim L, Wong K, Hayes-Klug J, Nalls A, Peyser D, Estevez V, Denkers N, Xu J, Osborn DA, Miller KV, Warren RJ, Brown DR, Chabalgoity JA, Hoover EA, Wisniewski T. Mucosal immunization with an attenuated Salmonella vaccine partially protects white-tailed deer from chronic wasting disease. Vaccine 2014; 33:726-33. [PMID: 25539804 DOI: 10.1016/j.vaccine.2014.11.035] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/13/2014] [Accepted: 11/19/2014] [Indexed: 10/24/2022]
Abstract
Prion disease is a unique category of illness, affecting both animals and humans, in which the underlying pathogenesis is related to a conformational change of a normal, self-protein called PrP(C) (C for cellular) to a pathological and infectious conformer known as PrP(Sc) (Sc for scrapie). Bovine spongiform encephalopathy (BSE), a prion disease believed to have arisen from feeding cattle with prion contaminated meat and bone meal products, crossed the species barrier to infect humans. Chronic wasting disease (CWD) infects large numbers of deer and elk, with the potential to infect humans. Currently no prionosis has an effective treatment. Previously, we have demonstrated we could prevent transmission of prions in a proportion of susceptible mice with a mucosal vaccine. In the current study, white-tailed deer were orally inoculated with attenuated Salmonella expressing PrP, while control deer were orally inoculated with vehicle attenuated Salmonella. Once a mucosal response was established, the vaccinated animals were boosted orally and locally by application of polymerized recombinant PrP onto the tonsils and rectal mucosa. The vaccinated and control animals were then challenged orally with CWD-infected brain homogenate. Three years post CWD oral challenge all control deer developed clinical CWD (median survival 602 days), while among the vaccinated there was a significant prolongation of the incubation period (median survival 909 days; p=0.012 by Weibull regression analysis) and one deer has remained CWD free both clinically and by RAMALT and tonsil biopsies. This negative vaccinate has the highest titers of IgA in saliva and systemic IgG against PrP. Western blots showed that immunoglobulins from this vaccinate react to PrP(CWD). We document the first partially successful vaccination for a prion disease in a species naturally at risk.
Collapse
Affiliation(s)
- Fernando Goñi
- Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States
| | - Candace K Mathiason
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Lucia Yim
- Laboratory for Vaccine Research, Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Kinlung Wong
- Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States
| | - Jeanette Hayes-Klug
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Amy Nalls
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Daniel Peyser
- Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States
| | - Veronica Estevez
- Laboratory for Vaccine Research, Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Nathaniel Denkers
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Jinfeng Xu
- Department of Population Health, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States
| | - David A Osborn
- Warnell School of Forestry and Natural Resources, University of Georgia, United States
| | - Karl V Miller
- Warnell School of Forestry and Natural Resources, University of Georgia, United States
| | - Robert J Warren
- Warnell School of Forestry and Natural Resources, University of Georgia, United States
| | - David R Brown
- Department of Biology and Biochemistry, University of Bath, UK
| | - Jose A Chabalgoity
- Laboratory for Vaccine Research, Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Edward A Hoover
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Thomas Wisniewski
- Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States; Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States; Department of Psychiatry, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States.
| |
Collapse
|
22
|
Prion protein-specific antibodies-development, modes of action and therapeutics application. Viruses 2014; 6:3719-37. [PMID: 25275428 PMCID: PMC4213558 DOI: 10.3390/v6103719] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 12/21/2022] Open
Abstract
Prion diseases or Transmissible Spongiform Encephalopathies (TSEs) are lethal neurodegenerative disorders involving the misfolding of the host encoded cellular prion protein, PrPC. This physiological form of the protein is expressed throughout the body, and it reaches the highest levels in the central nervous system where the pathology occurs. The conversion into the pathogenic isoform denoted as prion or PrPSc is the key event in prion disorders. Prominent candidates for the treatment of prion diseases are antibodies and their derivatives. Anti-PrPC antibodies are able to clear PrPSc from cell culture of infected cells. Furthermore, application of anti-PrPC antibodies suppresses prion replication in experimental animal models. Major drawbacks of immunotherapy are immune tolerance, the risks of neurotoxic side effects, limited ability of compounds to cross the blood-brain barrier and their unfavorable pharmacokinetic. The focus of this review is to recapitulate the current understanding of the molecular mechanisms for antibody mediated anti-prion activity. Although relevant for designing immunotherapeutic tools, the characterization of key antibody parameters shaping the molecular mechanism of the PrPC to PrPSc conversion remains elusive. Moreover, this review illustrates the various attempts towards the development of anti-PrP antibody compounds and discusses therapeutic candidates that modulate PrP expression.
Collapse
|
23
|
Development of a Multivalent, PrPSc-Specific Prion Vaccine through Rational Optimization of Three Disease-Specific Epitopes. Vaccine 2014; 32:1988-97. [DOI: 10.1016/j.vaccine.2014.01.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/23/2013] [Accepted: 01/14/2014] [Indexed: 11/21/2022]
|
24
|
Ludewigs H, Zuber C, Vana K, Nikles D, Zerr I, Weiss S. Therapeutic approaches for prion disorders. Expert Rev Anti Infect Ther 2014; 5:613-30. [PMID: 17678425 DOI: 10.1586/14787210.5.4.613] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Prion diseases are lethal for both humans and animals, and affected individuals die after several months following a rapid disease progression. Although researchers have attempted for decades to develop effective therapeutics for the therapy of human prion disorders, until now no efficient drug has been available on the market for transmissible spongiform encephalopathy (TSE) treatment or cure. Approximately 200 patients worldwide have died or suffer from variant Creutzfeldt-Jakob disease (CJD). Incidences for sporadic and familial CJD are approximately 1.5-2 per million per year and one per 10 million per year, respectively, in Europe. This review summarizes classical and modern trials for the development of effective anti-TSE drugs, introduces potential effective delivery systems, such as lentiviral and adeno-associated virus systems for antiprion components, including antibodies and siRNAs, and presents vaccination trials. Most of the antiprion drugs target prion protein PrP(c) and/or PrP(Sc). Alternative targets are receptors and coreceptors for PrP, that is, the 37/67-kDa laminin receptor and heparan sulfate proteoglycanes. We review clinical trials for the treatment of TSEs and describe hindrances and chances for a breakthrough in therapy of prion disorders.
Collapse
Affiliation(s)
- Heike Ludewigs
- Laboratorium für Molekulare Biologie, Genzentrum, Institut für Biochemie der LMU München, München, Germany.
| | | | | | | | | | | |
Collapse
|
25
|
Molecular dynamics studies on the NMR and X-ray structures of rabbit prion proteins. J Theor Biol 2013; 342:70-82. [PMID: 24184221 DOI: 10.1016/j.jtbi.2013.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 08/25/2013] [Accepted: 10/09/2013] [Indexed: 12/27/2022]
Abstract
Prion diseases, traditionally referred to as transmissible spongiform encephalopathies (TSEs), are invariably fatal and highly infectious neurodegenerative diseases that affect a wide variety of mammalian species, manifesting as scrapie in sheep and goats, bovine spongiform encephalopathy (BSE or mad-cow disease) in cattle, chronic wasting disease in deer and elk, and Creutzfeldt-Jakob diseases, Gerstmann-Sträussler-Scheinker syndrome, fatal familial insomnia, and kulu in humans, etc. These neurodegenerative diseases are caused by the conversion from a soluble normal cellular prion protein (PrP(C)) into insoluble abnormally folded infectious prions (PrP(Sc)), and the conversion of PrP(C) to PrP(Sc) is believed to involve conformational change from a predominantly α-helical protein to one rich in β-sheet structure. Such a conformational change may be amenable to study by molecular dynamics (MD) techniques. For rabbits, classical studies show that they have a low susceptibility to be infected by PrP(Sc), but recently it was reported that rabbit prions can be generated through saPMCA (serial automated Protein Misfolding Cyclic Amplification) in vitro and the rabbit prion is infectious and transmissible. In this paper, we first do a detailed survey on the research advances of rabbit prion protein (RaPrP) and then we perform MD simulations on the NMR and X-ray molecular structures of rabbit prion protein wild-type and mutants. The survey shows to us that rabbits were not challenged directly in vivo with other known prion strains and the saPMCA result did not pass the test of the known BSE strain of cattle. Thus, we might still look rabbits as a prion resistant species. MD results indicate that the three α-helices of the wild-type are stable under the neutral pH environment (but under low pH environment the three α-helices have been unfolded into β-sheets), and the three α-helices of the mutants (I214V and S173N) are unfolded into rich β-sheet structures under the same pH environment. In addition, we found an interesting result that the salt bridges such as ASP201-ARG155, ASP177-ARG163 contribute greatly to the structural stability of RaPrP.
Collapse
|
26
|
Wisniewski T, Goñi F. Could immunomodulation be used to prevent prion diseases? Expert Rev Anti Infect Ther 2012; 10:307-17. [PMID: 22397565 DOI: 10.1586/eri.11.177] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
All prion diseases are currently without effective treatment and are universally fatal. The underlying pathogenesis of prion diseases (prionoses) is related to an autocatalytic conformational conversion of PrP(C) (C for cellular) to a pathological and infectious conformer known as PrP(Sc) (Sc for scrapie) or PrP(Res) (Res for proteinase K resistant). The past experience with variant Creutzfeldt-Jakob disease, which originated from bovine spongiform encephalopathy, as well as the ongoing epidemic of chronic wasting disease has highlighted the necessity for effective prophylactic and/or therapeutic approaches. Human prionoses are most commonly sporadic, and hence therapy is primarily directed to stop progression; however, in animals the majority of prionoses are infectious and, as a result, the emphasis is on prevention of transmission. These infectious prionoses are most commonly acquired via the alimentary tract as a major portal of infectious agent entry, making mucosal immunization a potentially attractive method to produce a local immune response that can partially or completely prevent prion entry across the gut barrier, while at the same time producing a modulated systemic immunity that is unlikely to be associated with toxicity. A critical factor in any immunomodulatory methodology that targets a self-antigen is the need to delicately balance an effective humoral immune response with potential autoimmune inflammatory toxicity. The ongoing epidemic of chronic wasting disease affecting the USA and Korea, with the potential to spread to human populations, highlights the need for such immunomodulatory approaches.
Collapse
Affiliation(s)
- Thomas Wisniewski
- New York University School of Medicine, 560 First Avenue, New York, NY 10016, USA.
| | | |
Collapse
|
27
|
Hedlin P, Taschuk R, Potter A, Griebel P, Napper S. Detection and control of prion diseases in food animals. ISRN VETERINARY SCIENCE 2012; 2012:254739. [PMID: 23738120 PMCID: PMC3658581 DOI: 10.5402/2012/254739] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/22/2011] [Indexed: 12/14/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs), or prion diseases, represent a unique form of infectious disease based on misfolding of a self-protein (PrPC) into a pathological, infectious conformation (PrPSc). Prion diseases of food animals gained notoriety during the bovine spongiform encephalopathy (BSE) outbreak of the 1980s. In particular, disease transmission to humans, to the generation of a fatal, untreatable disease, elevated the perspective on livestock prion diseases from food production to food safety. While the immediate threat posed by BSE has been successfully addressed through surveillance and improved management practices, another prion disease is rapidly spreading. Chronic wasting disease (CWD), a prion disease of cervids, has been confirmed in wild and captive populations with devastating impact on the farmed cervid industries. Furthermore, the unabated spread of this disease through wild populations threatens a natural resource that is a source of considerable economic benefit and national pride. In a worst-case scenario, CWD may represent a zoonotic threat either through direct transmission via consumption of infected cervids or through a secondary food animal, such as cattle. This has energized efforts to understand prion diseases as well as to develop tools for disease detection, prevention, and management. Progress in each of these areas is discussed.
Collapse
Affiliation(s)
- Peter Hedlin
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK, Canada S7N 5E3 ; Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada S7N 5E3
| | | | | | | | | |
Collapse
|
28
|
|
29
|
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: 1.5] [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
|
30
|
Alexandrenne C, Wijkhuisen A, Dkhissi F, Hanoux V, Priam F, Allard B, Boquet D, Couraud JY. Electrotransfer of cDNA Coding for a Heterologous Prion Protein Generates Autoantibodies Against Native Murine Prion Protein in Wild-Type Mice. DNA Cell Biol 2010; 29:121-31. [DOI: 10.1089/dna.2009.0940] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Coralie Alexandrenne
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
| | - Anne Wijkhuisen
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
- UFR SdV, Paris Diderot University, Paris, France
| | - Fatima Dkhissi
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
- UFR SdV, Paris Diderot University, Paris, France
| | - Vincent Hanoux
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
- UFR SdV, Paris Diderot University, Paris, France
| | - Fabienne Priam
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
- UFR SdV, Paris Diderot University, Paris, France
| | - Bertrand Allard
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
| | - Didier Boquet
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
| | - Jean-Yves Couraud
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
- UFR SdV, Paris Diderot University, Paris, France
| |
Collapse
|
31
|
Gilch S, Bach C, Lutzny G, Vorberg I, Schätzl HM. Inhibition of cholesterol recycling impairs cellular PrP(Sc) propagation. Cell Mol Life Sci 2009; 66:3979-91. [PMID: 19823766 PMCID: PMC2777232 DOI: 10.1007/s00018-009-0158-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 09/14/2009] [Indexed: 12/19/2022]
Abstract
The infectious agent in prion diseases consists of an aberrantly folded isoform of the cellular prion protein (PrP(c)), termed PrP(Sc), which accumulates in brains of affected individuals. Studies on prion-infected cultured cells indicate that cellular cholesterol homeostasis influences PrP(Sc) propagation. Here, we demonstrate that the cellular PrP(Sc) content decreases upon accumulation of cholesterol in late endosomes, as induced by NPC-1 knock-down or treatment with U18666A. PrP(c) trafficking, lipid raft association, and membrane turnover are not significantly altered by such treatments. Cellular PrP(Sc) formation is not impaired, suggesting that PrP(Sc) degradation is increased by intracellular cholesterol accumulation. Interestingly, PrP(Sc) propagation in U18666A-treated cells was partially restored by overexpression of rab 9, which causes redistribution of cholesterol and possibly of PrP(Sc) to the trans-Golgi network. Surprisingly, rab 9 overexpression itself reduced cellular PrP(Sc) content, indicating that PrP(Sc) production is highly sensitive to alterations in dynamics of vesicle trafficking.
Collapse
Affiliation(s)
- Sabine Gilch
- Institute of Virology, Prion Research Group, Technische Universität München, Trogerstr. 30, 81675 Munich, Germany
| | - Christian Bach
- Institute of Virology, Prion Research Group, Technische Universität München, Trogerstr. 30, 81675 Munich, Germany
| | - Gloria Lutzny
- Institute of Virology, Prion Research Group, Technische Universität München, Trogerstr. 30, 81675 Munich, Germany
| | - Ina Vorberg
- Institute of Virology, Prion Research Group, Technische Universität München, Trogerstr. 30, 81675 Munich, Germany
| | - Hermann M. Schätzl
- Institute of Virology, Prion Research Group, Technische Universität München, Trogerstr. 30, 81675 Munich, Germany
| |
Collapse
|
32
|
Zhao L, Hou X, Ji R, Han C, Yu X, Hong T. Establishment of bovine prion peptide-based monoclonal antibodies for identifying bovine prion. ACTA ACUST UNITED AC 2009; 52:754-60. [PMID: 19727594 DOI: 10.1007/s11427-009-0100-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 05/04/2009] [Indexed: 10/20/2022]
Abstract
To obtain high titer monoclonal antibodies (McAbs) which can react with mammalian prion protein (PrP), Balb/C mice were immunized with bovine (Bo) PrP peptide (BoPrP 209-228 aa) coupled to keyhole limpet hemocyanin (KLH). The hybridoma cell lines secreting monoclonal antibodies against the peptide were established by cell fusion and cloning. The obtained McAbs were applied to detect recombinant human, bovine and hamster PrP, cellular prion protein (PrP(c)) in normal bovine brain and pathogenic scrapie prion protein (PrP(Sc)) accumulated in the medulla oblongata of bovine spongiform encephalopathy(BSE)specimen with Western blot and immunohistochemical detection, respectively. The current procedure might offer a simple, feasible method to raise high titer antibodies for studying biological features of PrP in mammals, as well as detection of transmissible spongiform encephalopathy (TSE) and diagnosis of BSE, in particular.
Collapse
Affiliation(s)
- Li Zhao
- Department of Laboratory Science of Microbiology, Shandong University, Jinan, 250012, China
| | | | | | | | | | | |
Collapse
|
33
|
Phosphorylation of prion protein at serine 43 induces prion protein conformational change. J Neurosci 2009; 29:8743-51. [PMID: 19587281 DOI: 10.1523/jneurosci.2294-09.2009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The cause of the conformational change of normal cellular prion protein (PrP) into its disease-associated form is unknown. Posttranslational modifications, such as glycosylation, acetylation, S-nitrosylation, and phosphorylation, are known to induce protein conformational changes. Here, we investigated whether phosphorylation could induce the conformational change of PrP because PrP contains several kinase motifs and has been found recently in the cytosol, in which kinases generally reside. Neuronal cyclin-dependent kinase 5 (Cdk5) phosphorylated recombinant PrP(23-231) at serine 43 (S43) in an in vitro kinase assay. Cdk5-phosphorylated PrP became proteinase K resistant, formed Congo Red-positive fibrils, and formed aggregates that were immunostained with anti-PrP and anti-phospho-PrP(S43) (anti-pPrP(S43)). pPrP(S43) was detected in PrP/Cdk5/p25 cotransfected N2a cells. Roscovitine inhibition of Cdk5 activity or transfection of N2a cells with mutant PrP S43A eliminated the anti-pPrP(S43)-immunopositive protein. Alkaline phosphatase-sensitive and proteinase K-resistant pPrP(S43) immunoreactivity was observed in scrapie-infected but not control-injected mice brains. These results raise the possibility that phosphorylation could represent a physiological mechanism of PrP conversion in vivo.
Collapse
|
34
|
Tayebi M, Collinge J, Hawke S. Unswitched immunoglobulin M response prolongs mouse survival in prion disease. J Gen Virol 2009; 90:777-782. [PMID: 19218226 DOI: 10.1099/vir.0.005041-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Several studies have failed to demonstrate the presence of immune responses to infectious prions during the course of prion disease, reflecting the identical primary structure of normal and disease-associated isoforms and the widespread expression of the normal cellular form of prion protein, PrP(C), leading to B- and/or T-cell tolerance of disease-associated isoforms and also possibly because antigen-presenting cells are unable to process the highly aggregated, detergent-insoluble, protease-resistant form, PrP(Sc). Under certain circumstances, PrP(Sc) can be revealed to the immune system in immunogenic form, and it has been shown previously that anti-PrP antibodies can be induced to prions immunoadsorbed to Dynabeads using specific anti-PrP monoclonal antibodies, even in PrP-sufficient mice. This study demonstrated in a murine scrapie model that PrP-Dynabeads effectively stimulated the immune system to produce anti-PrP IgM antibodies over prolonged periods after repeated immunization. It was also shown that these immune responses prolonged incubation times in murine scrapie.
Collapse
Affiliation(s)
- Mourad Tayebi
- Brain & Mind Research Institute, University of Sydney, 100 Mallett Street, Camperdown, NSW 2050, Australia.,Department of Pathology and Infectious Diseases, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK
| | - John Collinge
- MRC Prion Unit and Department of Neurodegenerative Disease, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Simon Hawke
- Brain & Mind Research Institute, University of Sydney, 100 Mallett Street, Camperdown, NSW 2050, Australia
| |
Collapse
|
35
|
Dendritic cell-mediated-immunization with xenogenic PrP and adenoviral vectors breaks tolerance and prolongs mice survival against experimental scrapie. PLoS One 2009; 4:e4917. [PMID: 19295917 PMCID: PMC2654673 DOI: 10.1371/journal.pone.0004917] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Accepted: 02/16/2009] [Indexed: 12/30/2022] Open
Abstract
In prion diseases, PrPc, a widely expressed protein, is transformed into a pathogenic form called PrPSc, which is in itself infectious. Antibodies directed against PrPc have been shown to inhibit PrPc to PrPSc conversion in vitro and protect in vivo from disease. Other effectors with potential to eliminate PrPSc-producing cells are cytotoxic T cells directed against PrP-derived peptides but their ability to protect or to induce deleterious autoimmune reactions is not known. The natural tolerance to PrPc makes difficult to raise efficient adaptive responses. To break tolerance, adenovirus (Ad) encoding human PrP (hPrP) or control Ad were administered to wild-type mice by direct injection or by transfer of Ad-transduced dendritic cells (DCs). Control Ad-transduced DCs from Tg650 mice overexpressing hPrP were also used for immunization. DC-mediated but not direct administration of AdhPrP elicited antibodies that bound to murine native PrPc. Frequencies of PrP-specific IFNγ-secreting T cells were low and in vivo lytic activity only targeted cells strongly expressing hPrP. Immunohistochemical analysis revealed that CD3+ T cell infiltration was similar in the brain of vaccinated and unvaccinated 139A-infected mice suggesting the absence of autoimmune reactions. Early splenic PrPSc replication was strongly inhibited ten weeks post infection and mean survival time prolonged from 209 days in untreated 139A-infected mice to 246 days in mice vaccinated with DCs expressing the hPrP. The efficacy appeared to be associated with antibody but not with cytotoxic cell-mediated PrP-specific responses.
Collapse
|
36
|
Crystal structure of human prion protein bound to a therapeutic antibody. Proc Natl Acad Sci U S A 2009; 106:2554-8. [PMID: 19204296 DOI: 10.1073/pnas.0809170106] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Prion infection is characterized by the conversion of host cellular prion protein (PrP(C)) into disease-related conformers (PrP(Sc)) and can be arrested in vivo by passive immunization with anti-PrP monoclonal antibodies. Here, we show that the ability of an antibody to cure prion-infected cells correlates with its binding affinity for PrP(C) rather than PrP(Sc). We have visualized this interaction at the molecular level by determining the crystal structure of human PrP bound to the Fab fragment of monoclonal antibody ICSM 18, which has the highest affinity for PrP(C) and the highest therapeutic potency in vitro and in vivo. In this crystal structure, human PrP is observed in its native PrP(C) conformation. Interactions between neighboring PrP molecules in the crystal structure are mediated by close homotypic contacts between residues at position 129 that lead to the formation of a 4-strand intermolecular beta-sheet. The importance of this residue in mediating protein-protein contact could explain the genetic susceptibility and prion strain selection determined by polymorphic residue 129 in human prion disease, one of the strongest common susceptibility polymorphisms known in any human disease.
Collapse
|
37
|
Gilch S, Krammer C, Schätzl HM. Targeting prion proteins in neurodegenerative disease. Expert Opin Biol Ther 2008; 8:923-40. [PMID: 18549323 DOI: 10.1517/14712598.8.7.923] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Spongiform neurodegeneration is the pathological hallmark of individuals suffering from prion disease. These disorders, whose manifestation is sporadic, familial or acquired by infection, are caused by accumulation of the aberrantly folded isoform of the cellular prion protein (PrP(c)), termed PrP(Sc). Although usually rare, prion disorders are inevitably fatal and transferrable by infection. OBJECTIVE Pathology is restricted to the central nervous system and premortem diagnosis is usually not possible. Yet, promising approaches towards developing therapeutic regimens have been made recently. METHODS The biology of prion proteins and current models of neurotoxicity are discussed and prophylactic and therapeutic concepts are introduced. RESULTS/CONCLUSIONS Although various promising drug candidates with antiprion activity have been identified, this proof-of-concept cannot be transferred into translational medicine yet.
Collapse
Affiliation(s)
- Sabine Gilch
- Technische Universität München, Institute of Virology, Prion Research Group, Trogerstreet 30, 81675 Munich, Germany
| | | | | |
Collapse
|
38
|
Song CH, Furuoka H, Kim CL, Ogino M, Suzuki A, Hasebe R, Horiuchi M. Effect of intraventricular infusion of anti-prion protein monoclonal antibodies on disease progression in prion-infected mice. J Gen Virol 2008; 89:1533-1544. [PMID: 18474571 DOI: 10.1099/vir.0.83578-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
It is well known that anti-prion protein (PrP) monoclonal antibodies (mAbs) inhibit abnormal isoform PrP (PrPSc) formation in cell culture. Additionally, passive immunization of anti-PrP mAbs protects the animals from prion infection via peripheral challenge when mAbs are administered simultaneously or soon after prion inoculation. Thus, anti-PrP mAbs are candidates for the treatment of prion diseases. However, the effects of mAbs on disease progression in the middle and late stages of the disease remain unclear. This study carried out intraventricular infusion of mAbs into prion-infected mice before and after clinical onset to assess their ability to delay disease progression. A 4-week infusion of anti-PrP mAbs initiated at 120 days post-inoculation (p.i.), which is just after clinical onset, reduced PrPSc levels to 70-80 % of those found in mice treated with a negative-control mAb. Spongiform changes, microglial activation and astrogliosis in the hippocampus and thalamus appeared milder in mice treated with anti-PrP mAbs than in those treated with a negative-control mAb. Treatment with anti-PrP mAb prolonged the survival of mice infected with Chandler or Obihiro strain when infusion was initiated at 60 days p.i., at which point PrPSc is detectable in the brain. In contrast, infusion initiated after clinical onset prolonged the survival time by about 8 % only in mice infected with the Chandler strain. Although the effects on survival varied for different prion strains, the anti-PrP mAb could partly prevent disease progression, even after clinical onset, suggesting immunotherapy as a candidate for treatment of prion diseases.
Collapse
Affiliation(s)
- Chang-Hyun Song
- Laboratory of Prion Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Hidefumi Furuoka
- Department of Pathobiological Science, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro 080-8555, Japan
| | - Chan-Lan Kim
- Laboratory of Prion Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Michiko Ogino
- Laboratory of Prion Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Akio Suzuki
- Laboratory of Prion Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Rie Hasebe
- Laboratory of Prion Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Motohiro Horiuchi
- Laboratory of Prion Diseases, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| |
Collapse
|
39
|
Müller-Schiffmann A, Korth C. Vaccine approaches to prevent and treat prion infection : progress and challenges. BioDrugs 2008; 22:45-52. [PMID: 18215090 DOI: 10.2165/00063030-200822010-00005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Prion diseases are transmissible neurodegenerative diseases of humans and animals. The prion agent consists of a misfolded protein, PrPSc (prion protein, scrapie form), of a glycosylphosphatidylinositol-anchored host protein, PrPC (PrP cellular form) of unknown function. During prion replication, PrPSc induces host PrPC to adopt its pathogenic conformation. Some PrPSc may aggregate to microscopically visible, extracellular prion plaques that stain for amyloid. The development of antiprion vaccines presents some challenges. While there is strong self-tolerance to an endogenous antibody response to PrPC and PrPSc, highly potent monoclonal antibodies (mAbs) have been raised in mice in which the prion protein gene has been deleted by gene targeting. These mAbs have been demonstrated to be antiprion-active in permanently scrapie-infected neuroblastoma (ScN2a) cells, primarily when bound to one of four epitopes (the octarepeat region, the region around codons 90-110, helix 1 region codons 145-160, and the extreme C-terminal codons 210-220). The mAbs directed against codon regions 90-110 or 145-160 are also antiprion-active in vivo, but only after intraperitoneal infection with prions, not intracerebral infection, suggesting their blood-brain barrier (BBB) impermeability. The challenge will be to make antibodies, or recombinant derivatives thereof, BBB permeable; this is preferably achieved by monovalent antibody fragments since divalent ones were found to be neurotoxic. Self-tolerance of wild-type animals to PrP immunizations was found to be of extrathymic origin. Even though antibodies raised in wild-type mice were found to display antiprion activity in ScN2a cells, these mice did not have significant extensions of incubation times when challenged intraperitoneally with prions. A general low affinity of these antibody responses to native surface-bound PrPC may account for this. Since wild-type mice were found to develop sufficient T-cell responses to codon regions 145-160 and 210-220, we believe that there is a theoretical chance of a successful vaccination therapy. The influence of the way the immunogen is presented has already been shown to be of major importance for the ensuing immune response, in that presentation of PrP with CpG oligodeoxynucleotides as adjuvant or viral packaging improved antibody responses. Major progress for active immunizations may therefore be expected in this field. Eradication programs will be one of the most important uses of active immunization protocols. For this purpose, vaccines will have to be inexpensive, easy to handle, and effective. In the short term, passive immunizations will likely be most promising for therapy of prion disease, including for human medical interventions. Active immunization protocols are less likely to succeed quickly, and will take years if not decades to be validated for domestic or free-ranging animals.
Collapse
|
40
|
Aguib Y, Gilch S, Krammer C, Ertmer A, Groschup MH, Schätzl HM. Neuroendocrine cultured cells counteract persistent prion infection by down-regulation of PrPc. Mol Cell Neurosci 2008; 38:98-109. [DOI: 10.1016/j.mcn.2008.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2007] [Revised: 01/30/2008] [Accepted: 02/13/2008] [Indexed: 11/25/2022] Open
|
41
|
Heiseke A, Schöbel S, Lichtenthaler SF, Vorberg I, Groschup MH, Kretzschmar H, Schätzl HM, Nunziante M. The novel sorting nexin SNX33 interferes with cellular PrP formation by modulation of PrP shedding. Traffic 2008; 9:1116-29. [PMID: 18419754 DOI: 10.1111/j.1600-0854.2008.00750.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The cellular prion protein (PrP(c)) is a glycosyl-phosphatidylinositol (GPI)-anchored protein trafficking in the secretory and endocytic pathway and localized mainly at the plasma membrane. Conversion of PrP(c) into its pathogenic isoform PrP(Sc) is associated with pathogenesis and transmission of prion diseases. Intramolecular cleavage in the middle, the extreme C-terminal part or within the GPI anchor and shedding of PrP(c) modulate this conversion process by reducing the substrate for prion formation. These phenomena provide similarities with the processing of amyloid precursor protein in Alzheimer's disease. Sorting nexins are a family of proteins with important functions in protein trafficking. In this study, we investigated the role of the newly described sorting nexin 33 (SNX33) in trafficking and processing of PrP(c). We found that overexpression of SNX33 in neuronal and non-neuronal cell lines resulted in increased shedding of full-length PrP(c) from the plasma membrane and modulated the rate of PrP(c) endocytosis. This was paralleled by reduction of PrP(Sc) formation in persistently and newly infected cells. Using deletion mutants, we demonstrate that production of PrP fragment N1 is not influenced by SNX33. Our data provide new insights into the cellular mechanisms of PrP(c) shedding and show how this can affect cellular PrP(Sc) conversion.
Collapse
Affiliation(s)
- Andreas Heiseke
- Institute of Virology, Technical University of Munich, Trogerstr. 30, 81675 Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Hooper NM, Turner AJ. A new take on prions: preventing Alzheimer's disease. Trends Biochem Sci 2008; 33:151-5. [DOI: 10.1016/j.tibs.2008.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 01/11/2008] [Accepted: 01/16/2008] [Indexed: 12/31/2022]
|
43
|
Goñi F, Prelli F, Schreiber F, Scholtzova H, Chung E, Kascsak R, Brown DR, Sigurdsson EM, Chabalgoity JA, Wisniewski T. High titers of mucosal and systemic anti-PrP antibodies abrogate oral prion infection in mucosal-vaccinated mice. Neuroscience 2008; 153:679-86. [PMID: 18407424 DOI: 10.1016/j.neuroscience.2008.02.051] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Revised: 02/02/2008] [Accepted: 02/15/2008] [Indexed: 12/28/2022]
Abstract
Significant outbreaks of prion disease linked to oral exposure of the prion agent have occurred in animal and human populations. These disorders are associated with a conformational change of a normal protein, PrP(C) (C for cellular), to a toxic and infectious form, PrP(Sc) (Sc for scrapie). None of the prionoses currently have an effective treatment. Some forms of prion disease are thought to be spread by oral ingestion of PrP(Sc), such as chronic wasting disease and variant Creutzfeldt-Jakob disease. Attempts to obtain an active immunization in wild-type animals have been hampered by auto-tolerance to PrP and potential toxicity. Previously, we demonstrated that it is possible to overcome tolerance and obtain a specific anti-PrP antibody response by oral inoculation of the PrP protein expressed in an attenuated Salmonella vector. This past study showed that 30% of vaccinated animals were free of disease more than 350 days post-challenge. In the current study we have both optimized the vaccination protocol and divided the vaccinated mice into low and high immune responder groups prior to oral challenge with PrP(Sc) scrapie strain 139A. These methodological refinements led to a significantly improved therapeutic response. 100% of mice with a high mucosal anti-PrP titer immunoglobulin (Ig) A and a high systemic IgG titer, prior to challenge, remained without symptoms of PrP infection at 400 days (log-rank test P<0.0001 versus sham controls). The brains from these surviving clinically asymptomatic mice were free of PrP(Sc) infection by Western blot and histological examination. These promising findings suggest that effective mucosal vaccination is a feasible and useful method for overcoming tolerance to PrP and preventing prion infection via an oral route.
Collapse
Affiliation(s)
- F Goñi
- Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Vilette D. Cell models of prion infection. Vet Res 2007; 39:10. [PMID: 18073097 DOI: 10.1051/vetres:2007049] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Accepted: 09/24/2007] [Indexed: 11/14/2022] Open
Abstract
Due to recent renewal of interest and concerns in prion diseases, a number of cell systems permissive to prion multiplication have been generated in the last years. These include established cell lines, neuronal stem cells and primary neuronal cultures. While most of these models are permissive to experimental, mouse-adapted strains of prions, the propagation of natural field isolates from sheep scrapie and chronic wasting disease has been recently achieved. These models have improved our knowledge on the molecular and cellular events controlling the conversion of the PrP(C) protein into abnormal isoforms and on the cell-to-cell spreading of prions. Infected cultured cells will also facilitate investigations on the molecular basis of strain identity and on the mechanisms that lead to neurodegeneration. The ongoing development of new cell models with improved characteristics will certainly be useful for a number of unanswered critical issues in the prion field.
Collapse
Affiliation(s)
- Didier Vilette
- Unité Mixte de Recherche 1225, INRA, ENVT, 31000 Toulouse, France.
| |
Collapse
|
45
|
Kaiser-Schulz G, Heit A, Quintanilla-Martinez L, Hammerschmidt F, Hess S, Jennen L, Rezaei H, Wagner H, Schätzl HM. Polylactide-coglycolide microspheres co-encapsulating recombinant tandem prion protein with CpG-oligonucleotide break self-tolerance to prion protein in wild-type mice and induce CD4 and CD8 T cell responses. THE JOURNAL OF IMMUNOLOGY 2007; 179:2797-807. [PMID: 17709493 DOI: 10.4049/jimmunol.179.5.2797] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Prion diseases are fatal neurodegenerative diseases that are characterized by the conformational conversion of the normal, mainly alpha-helical cellular prion protein (PrP) into the abnormal beta-sheet-rich infectious isoform (PrP(Sc)). The immune system neither shows reaction against cellular PrP nor PrP(Sc), most likely due to profound self-tolerance. In previous studies, we were able to partly overcome self-tolerance using recombinantly expressed dimeric PrP (tandem PrP (tPrP)), in association with different adjuvants. Proof of principle for antiprion efficacy was obtained in vitro and in vivo. In this study, we demonstrate the induction of a specific Th1 T cell response in wild-type mice immunized with tPrP and CpG-oligonucleotide (ODN). Biochemical influences such as refolding conditions, ionic strength, pH, and interaction with CpG-ODN affected antigenic structure and thus improved immunogenicity. Furthermore, s.c. immunization with tPrP and CpG-ODN co-encapsulated in biodegradable polylactide-coglycolide microspheres (PLGA-MS) enhanced CD4 T cell responses and, more prominent, the induction of CD8 T cells. In this vaccination protocol, PLGA-MS function as endosomal delivery device of Ag plus CpG-ODN to macrophages and dendritic cells. In contrast, PLGA-MS-based DNA vaccination approaches with a tPrP construct generated poor humoral and T cell responses. Our data show that prophylactic and therapeutic immunization approaches against prion infections might be feasible using tPrP Ag and CpG-ODN adjuvant without detectable side effects.
Collapse
Affiliation(s)
- Gunnar Kaiser-Schulz
- Institute of Virology, Prion Research Group, Technical University of Munich, Trogerstrasse 30, 81675 Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cernilec M, Vranac T, Hafner-Bratkovic I, Koren S, Venturini AC, Popović M, Juntes P, Serbec VC. Identification of an epitope on the recombinant bovine PrP that is able to elicit a prominent immune response in wild-type mice. Immunol Lett 2007; 113:29-39. [PMID: 17884181 DOI: 10.1016/j.imlet.2007.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Revised: 06/22/2007] [Accepted: 07/18/2007] [Indexed: 11/22/2022]
Abstract
The main cause for the development of transmissible spongiform encephalopathies (TSE) is the conformational change of prion protein from the normal cellular isoform (PrP(C)) into the abnormal isoform, named prion (PrP(Sc)). The two isoforms have the same primary structure, and with PrP being highly conserved among different species, no immune response to PrP(Sc) has been observed in infected humans or other mammals so far. The problem of inducing immune response was encountered when producing monoclonal antibodies against PrP, therefore mice lacking a functional Prnp gene were predominantly used for the immunization. In the present paper we report that by immunizing wild-type BALB/c mice with chemically unmodified recombinant bovine PrP a potent humoral immune response was achieved. Furthermore, we were able to isolate the monoclonal antibody (mAb) E12/2 and few other mAbs, all reacting specifically with bovine and human PrP, but not with PrP from several other mammals. The epitope of mAb E12/2 is located at the C-terminal end of helix 1, with His155 being crucial for binding. It has been proven that mAb E12/2 is useful for human and bovine TSE research as well as for diagnostics. Our results show that there are sufficient structural differences between mouse and bovine PrP to provoke a prominent humoral immune response.
Collapse
Affiliation(s)
- Maja Cernilec
- Blood Transfusion Centre of Slovenia, Slajmerjeva 6, 1000 Ljubljana, Slovenia
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Simoneau S, Rezaei H, Salès N, Kaiser-Schulz G, Lefebvre-Roque M, Vidal C, Fournier JG, Comte J, Wopfner F, Grosclaude J, Schätzl H, Lasmézas CI. In vitro and in vivo neurotoxicity of prion protein oligomers. PLoS Pathog 2007; 3:e125. [PMID: 17784787 PMCID: PMC1959381 DOI: 10.1371/journal.ppat.0030125] [Citation(s) in RCA: 184] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Accepted: 07/10/2007] [Indexed: 11/18/2022] Open
Abstract
The mechanisms underlying prion-linked neurodegeneration remain to be elucidated, despite several recent advances in this field. Herein, we show that soluble, low molecular weight oligomers of the full-length prion protein (PrP), which possess characteristics of PrP to PrPsc conversion intermediates such as partial protease resistance, are neurotoxic in vitro on primary cultures of neurons and in vivo after subcortical stereotaxic injection. Monomeric PrP was not toxic. Insoluble, fibrillar forms of PrP exhibited no toxicity in vitro and were less toxic than their oligomeric counterparts in vivo. The toxicity was independent of PrP expression in the neurons both in vitro and in vivo for the PrP oligomers and in vivo for the PrP fibrils. Rescue experiments with antibodies showed that the exposure of the hydrophobic stretch of PrP at the oligomeric surface was necessary for toxicity. This study identifies toxic PrP species in vivo. It shows that PrP-induced neurodegeneration shares common mechanisms with other brain amyloidoses like Alzheimer disease and opens new avenues for neuroprotective intervention strategies of prion diseases targeting PrP oligomers.
Collapse
Affiliation(s)
- Steve Simoneau
- Commissariat à l'Energie Atomique, Fontenay-aux-Roses, France
| | - Human Rezaei
- Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - Nicole Salès
- Department of Infectology, The Scripps Research Institute, Jupiter, Florida, United States of America
| | | | - Maxime Lefebvre-Roque
- Commissariat à l'Energie Atomique, Fontenay-aux-Roses, France
- Department of Infectology, The Scripps Research Institute, Jupiter, Florida, United States of America
| | | | | | - Julien Comte
- Commissariat à l'Energie Atomique, Fontenay-aux-Roses, France
| | - Franziska Wopfner
- Institute of Virology, Technical University of Munich, Munich, Germany
| | - Jeanne Grosclaude
- Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - Hermann Schätzl
- Institute of Virology, Technical University of Munich, Munich, Germany
| | - Corinne Ida Lasmézas
- Department of Infectology, The Scripps Research Institute, Jupiter, Florida, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
48
|
Tayebi M, Bate C, Hawke S, Williams A. A role for B lymphocytes in anti-infective prion therapies? Expert Rev Anti Infect Ther 2007; 5:631-8. [PMID: 17678426 DOI: 10.1586/14787210.5.4.631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The deposition of proteins in the form of amyloid fibrils and plaques is the characteristic feature of a number of neurodegenerative conditions affecting the nervous system. These disorders include prion and Alzheimer's diseases and are of enormous importance for public health. It has become apparent over the last 20 years that specificity and application in prion diseases' diagnostic and therapeutic situations are the most important considerations in designing strategies for the generation of antiprion antibodies. Specific antiprion therapeutics have been suggested and the establishment of the 'proof-of-principle' that the use of epitope-specific antiprion antibodies leads to indefinite delay of disease onset, has increased momentum for its use, although caution should be exerted prior to the application of new therapeutic strategies in a clinical set up. Furthermore, in vivo stimulation of immune-competent cells to specifically recognize and neutralize the abnormally folded isoform should also be pursued.
Collapse
Affiliation(s)
- Mourad Tayebi
- Department of Pathology and Infectious Diseases, The Royal Veterinary College, North Mymms, Hatfield, Hertfordshire, UK.
| | | | | | | |
Collapse
|
49
|
Gilch S, Kehler C, Schätzl HM. Peptide Aptamers Expressed in the Secretory Pathway Interfere with Cellular PrPSc Formation. J Mol Biol 2007; 371:362-73. [PMID: 17574575 DOI: 10.1016/j.jmb.2007.05.052] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2007] [Revised: 05/11/2007] [Accepted: 05/17/2007] [Indexed: 11/22/2022]
Abstract
Prion diseases are rare and obligatory fatal neurodegenerative disorders caused by the accumulation of a misfolded isoform (PrPSc) of the host-encoded prion protein (PrPc). Prophylactic and therapeutic regimens against prion diseases are very limited. To extend such strategies we selected peptide aptamers binding to PrP from a combinatorial peptide library presented on the Escherichia coli thioredoxin A (trxA) protein as a scaffold. In a yeast two-hybrid screen employing full-length murine PrP (aa 23-231) as a bait we identified three peptide aptamers that reproducibly bind to PrP. Treatment of prion-infected cells with recombinantly expressed aptamers added to the culture medium abolished PrPSc conversion with an IC50 between 350 and 700 nM. For expression in eukaryotic cells, peptide aptamers were fused to an N-terminal signal peptide for entry of the secretory pathway. The C terminus was modified by a glycosyl-phosphatidyl-inositol-(GPI) anchoring signal, a KDEL retention motif and the transmembrane and cytosolic domain of LAMP-I, respectively. These peptide aptamers retained their binding properties to PrPc and, depending on peptide sequence and C-terminal modification, interfered with endogenous PrPSc conversion upon expression in prion-infected cells. Notably, infection of cell cultures could be prevented by expression of KDEL peptide aptamers. For the first time, we show that trxA-based peptide aptamers can be targeted to the secretory pathway, thereby not losing the affinity for their target protein. Beside their inhibitory effect on prion conversion, these molecules could be used as fundament for rational drug design.
Collapse
Affiliation(s)
- Sabine Gilch
- Institute of Virology, Prion Research Group, Technical University Munich, Trogerstr. 30, 81675 Munich, Germany
| | | | | |
Collapse
|
50
|
Lefebvre-Roque M, Kremmer E, Gilch S, Zou WQ, Féraudet C, Gilles CM, Salès N, Grassi J, Gambetti P, Baron T, Schätzl H, Lasmézas CI. Toxic effects of intracerebral PrP antibody administration during the course of BSE infection in mice. Prion 2007; 1:198-206. [PMID: 19164902 DOI: 10.4161/pri.1.3.4870] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The absence of specific immune response is a hallmark of prion diseases. However, in vitro and in vivo experiments have provided evidence that an anti-PrP humoral response could have beneficial effects. Prophylactic passive immunization performed at the time of infection delayed or prevented disease. Nonetheless, the potential therapeutic effect of PrP antibodies administered shortly before the clinical signs has never been tested in vivo. Moreover, a recent study showed the potential toxicity of PrP antibodies administered intracerebrally. We aimed at evaluating the effect of a prolonged intracerebral anti-PrP antibody administration at the time of neuroinvasion in BSE infected Tg20 mice. Unexpectedly, despite a good penetration of the antibodies in the brain parenchyma, the treatment was not protective against the development of BSE. Instead, it led to an extensive neuronal loss, strong astrogliosis and microglial activation. Since this effect was observed after injection of anti-PrP antibodies as whole IgGs, F(ab')(2) or Fab fragments, the toxicity was directly related to the ability of the antibodies to recognize native PrP and to the intracerebral concentration achieved, and not to the Fc portion or the divalence of the antibodies. This experiment shows that a prolonged treatment with anti-PrP antibodies by the intracerebral route can induce severe side-effects and calls for caution with regard to the use of similar approaches for late therapeutic interventions in humans.
Collapse
Affiliation(s)
- Maxime Lefebvre-Roque
- Department of Infectology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|