101
|
Abstract
Prion disease research has opened up the "black-box" of neurodegeneration, defining a key role for protein misfolding wherein a predominantly alpha-helical precursor protein, PrP (C), is converted to a disease-associated, β-sheet enriched isoform called PrP (Sc). In Alzheimer disease (AD) the Aβ peptide derived from the β-amyloid precuror protein APP folds in β-sheet amyloid. Early thoughts along the lines of overlap may have been on target, (1) but were eclipsed by a simultaneous (but now anachronistic) controversy over the role of PrP (Sc) in prion diseases. (2) (,) (3) Nonetheless, as prion diseases such as Creutzfeldt-Jakob Disease (CJD) are themselves rare and can include an overt infectious mode of transmission, and as familial prion diseases and familial AD involve different genes, an observer might reasonably have concluded that prion research could occasionally catalyze ideas in AD, but could never provide concrete overlaps at the mechanistic level. Surprisingly, albeit a decade or three down the road, several prion/AD commonalities can be found within the contemporary literature. One important prion/AD overlap concerns seeded spread of Aβ aggregates by intracerebral inoculation much like prions, (4) and, with a neuron-to-neuron 'spreading' also reported for pathologic forms of other misfolded proteins, Tau (5) (,) (6) and α-synuclein in the case of Parkinson Disease. (7) (,) (8) The concept of seeded spread has been discussed extensively elsewhere, sometimes under the rubric of "prionoids" (9), and lies outside the scope of this particular review where we will focus upon PrP (C). From this point the story can now be subdivided into four strands of investigation: (1) pathologic effects of Aβ can be mediated by binding to PrP (C), (10) (2) the positioning of endoproteolytic processing events of APP by pathologic (β-cleavage + γ-cleavage) and non-pathologic (α-cleavage + γ-cleavage) secretase pathways is paralleled by seemingly analogous α- and β-like cleavage of PrP (C) (Fig. 1) (3) similar lipid raft environments for PrP (C) and APP processing machinery, (11) (-) (13) and perhaps in consequence, overlaps in repertoire of the PrP (C) and APP protein interactors ("interactomes"), (14) (,) (15) and (4) rare kindreds with mixed AD and prion pathologies. (16) Here we discuss confounds, consensus and conflict associated with parameters that apply to these experimental settings.
Collapse
Affiliation(s)
- David Westaway
- Department of Medicine (Neurology); University of Alberta; Edmonton, AB Canada
- Centre for Prions and Protein Folding Diseases; University of Alberta; Edmonton, AB Canada
- Department of Biochemistry; University of Alberta; Edmonton, AB Canada
| | - Jack H. Jhamandas
- Department of Medicine (Neurology); University of Alberta; Edmonton, AB Canada
| |
Collapse
|
102
|
Moda F, Vimercati C, Campagnani I, Ruggerone M, Giaccone G, Morbin M, Zentilin L, Giacca M, Zucca I, Legname G, Tagliavini F. Brain delivery of AAV9 expressing an anti-PrP monovalent antibody delays prion disease in mice. Prion 2012; 6:383-90. [PMID: 22842862 DOI: 10.4161/pri.20197] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Prion diseases are caused by a conformational modification of the cellular prion protein (PrP (C)) into disease-specific forms, termed PrP (Sc), that have the ability to interact with PrP (C) promoting its conversion to PrP (Sc). In vitro studies demonstrated that anti-PrP antibodies inhibit this process. In particular, the single chain variable fragment D18 antibody (scFvD18) showed high efficiency in curing chronically prion-infected cells. This molecule binds the PrP (C) region involved in the interaction with PrP (Sc) thus halting further prion formation. These findings prompted us to test the efficiency of scFvD18 in vivo. A recombinant Adeno-Associated Viral vector serotype 9 was used to deliver scFvD18 to the brain of mice that were subsequently infected by intraperitoneal route with the mouse-adapted scrapie strain RML. We found that the treatment was safe, prolonged the incubation time of scrapie-infected animals and decreased the burden of total proteinase-resistant PrP (Sc) in the brain, suggesting that scFvD18 interferes with prion replication in vivo. This approach is relevant for designing new therapeutic strategies for prion diseases and other disorders characterized by protein misfolding.
Collapse
Affiliation(s)
- Fabio Moda
- Division of Neuropathology and Neurology 5, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Alzheimer's disease Aβ assemblies mediating rapid disruption of synaptic plasticity and memory. Mol Brain 2012; 5:25. [PMID: 22805374 PMCID: PMC3502131 DOI: 10.1186/1756-6606-5-25] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 06/22/2012] [Indexed: 01/24/2023] Open
Abstract
Alzheimer’s disease (AD) is characterized by episodic memory impairment that often precedes clinical diagnosis by many years. Probing the mechanisms of such impairment may provide much needed means of diagnosis and therapeutic intervention at an early, pre-dementia, stage. Prior to the onset of significant neurodegeneration, the structural and functional integrity of synapses in mnemonic circuitry is severely compromised in the presence of amyloidosis. This review examines recent evidence evaluating the role of amyloid-ß protein (Aβ) in causing rapid disruption of synaptic plasticity and memory impairment. We evaluate the relative importance of different sizes and conformations of Aβ, including monomer, oligomer, protofibril and fibril. We pay particular attention to recent controversies over the relevance to the pathophysiology of AD of different water soluble Aβ aggregates and the importance of cellular prion protein in mediating their effects. Current data are consistent with the view that both low-n oligomers and larger soluble assemblies present in AD brain, some of them via a direct interaction with cellular prion protein, cause synaptic memory failure. At the two extremes of aggregation, monomers and fibrils appear to act in vivo both as sources and sinks of certain metastable conformations of soluble aggregates that powerfully disrupt synaptic plasticity. The same principle appears to apply to other synaptotoxic amyloidogenic proteins including tau, α-synuclein and prion protein.
Collapse
|
104
|
Llorens F, Del Río JA. Unraveling the neuroprotective mechanisms of PrP (C) in excitotoxicity. Prion 2012; 6:245-51. [PMID: 22437735 DOI: 10.4161/pri.19639] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Knowledge of the natural roles of cellular prion protein (PrP (C) ) is essential to an understanding of the molecular basis of prion pathologies. This GPI-anchored protein has been described in synaptic contacts, and loss of its synaptic function in complex systems may contribute to the synaptic loss and neuronal degeneration observed in prionopathy. In addition, Prnp knockout mice show enhanced susceptibility to several excitotoxic insults, GABAA receptor-mediated fast inhibition was weakened, LTP was modified and cellular stress increased. Although little is known about how PrP (C) exerts its function at the synapse or the downstream events leading to PrP (C) -mediated neuroprotection against excitotoxic insults, PrP (C) has recently been reported to interact with two glutamate receptor subunits (NR2D and GluR6/7). In both cases the presence of PrP (C) blocks the neurotoxicity induced by NMDA and Kainate respectively. Furthermore, signals for seizure and neuronal cell death in response to Kainate in Prnp knockout mouse are associated with JNK3 activity, through enhancing the interaction of GluR6 with PSD-95. In combination with previous data, these results shed light on the molecular mechanisms behind the role of PrP (C) in excitotoxicity. Future experimental approaches are suggested and discussed.
Collapse
Affiliation(s)
- Franc Llorens
- Molecular and Cellular Neurobiotechnology Group, Institut de Bioenginyeria de Catalunya (IBEC), Parc Científic de Barcelona, Barcelona, Spain.
| | | |
Collapse
|
105
|
Šnajder M, Vilfan T, Černilec M, Rupreht R, Popović M, Juntes P, Šerbec VČ, Ulrih NP. Enzymatic degradation of PrPSc by a protease secreted from Aeropyrum pernix K1. PLoS One 2012; 7:e39548. [PMID: 22761822 PMCID: PMC3386259 DOI: 10.1371/journal.pone.0039548] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 05/22/2012] [Indexed: 11/18/2022] Open
Abstract
Background An R30 fraction from the growth medium of Aeropyrum pernix was analyzed for the protease that can digest the pathological prion protein isoform (PrPSc) from different species (human, bovine, deer and mouse). Methodology/Principal Findings Degradation of the PrPSc isoform by the R30 fraction and the purified protease was evaluated using the 6H4 anti-PrP monoclonal antibody. Fragments from the N-terminal and C-terminal of PrPSc were also monitored by Western blotting using the EB8 anti-PrP monoclonal antibody, and by dot blotting using the C7/5 anti-PrP monoclonal antibody, respectively. For detection of smaller peptides from incomplete digestion of PrPSc, the EB8 monoclonal antibody was used after precipitation with sodium phosphotungstate. Characterization of the purified active protease from the R30 fraction was achieved, through purification by fast protein liquid chromatography, and identification by tandem mass spectrometry the serine metalloprotease pernisine. SDS-PAGE and zymography show the purified pernisine plus its proregion with a molecular weight of ca. 45 kDa, and the mature purified pernisine as ca. 23 kDa. The purified pernisine was active between 58°C and 99°C, and between pH 3.5 and 8.0. The temperature and pH optima of the enzymatic activity of the purified pernisine in the presence of 1 mM CaCl2 were 105°C ±0.5°C and pH 6.5±0.2, respectively. Conclusions/Significance Our study has identified and characterized pernisine as a thermostable serine metalloprotease that is secreted from A. pernix and that can digest the pathological prion protein PrPSc.
Collapse
Affiliation(s)
- Marko Šnajder
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Vilfan
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Černilec
- Blood Transfusion Center of Slovenia, Ljubljana, Slovenia
| | - Ruth Rupreht
- Blood Transfusion Center of Slovenia, Ljubljana, Slovenia
| | - Mara Popović
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Polona Juntes
- Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | | | - Nataša Poklar Ulrih
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins (CipKeBiP), Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|
106
|
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
|
107
|
Abstract
Prion science has been on a rollercoaster for two decades. In the mid 1990s, the specter of mad cow disease (bovine spongiform encephalopathy, BSE) provoked an unprecedented public scare that was first precipitated by the realization that this animal prion disease could be transmitted to humans and then rekindled by the evidence that BSE-infected humans could pass on the infection through blood transfusions. Along with the gradual disappearance of BSE, the interest in prions has waned with the general public, funding agencies and prospective PhD students. In the past few years, however, a bewildering variety of diseases have been found to share features with prion infections, including cell-to-cell transmission. Here we review these developments and summarize those open questions that we currently deem most interesting in prion biology: how do prions damage their hosts, and how do hosts attempt to neutralize invading prions?
Collapse
|
108
|
Cronier S, Carimalo J, Schaeffer B, Jaumain E, Béringue V, Miquel MC, Laude H, Peyrin JM. Endogenous prion protein conversion is required for prion-induced neuritic alterations and neuronal death. FASEB J 2012; 26:3854-61. [PMID: 22661006 DOI: 10.1096/fj.11-201772] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Prions cause fatal neurodegenerative conditions and result from the conversion of host-encoded cellular prion protein (PrP(C)) into abnormally folded scrapie PrP (PrP(Sc)). Prions can propagate both in neurons and astrocytes, yet neurotoxicity mechanisms remain unclear. Recently, PrP(C) was proposed to mediate neurotoxic signaling of β-sheet-rich PrP and non-PrP conformers independently of conversion. To investigate the role of astrocytes and neuronal PrP(C) in prion-induced neurodegeneration, we set up neuron and astrocyte primary cocultures derived from PrP transgenic mice. In this system, prion-infected astrocytes delivered ovine PrP(Sc) to neurons lacking PrP(C) (prion-resistant), or expressing a PrP(C) convertible (sheep) or not (mouse, human). We show that interaction between neuronal PrP(C) and exogenous PrP(Sc) was not sufficient to induce neuronal death but that efficient PrP(C) conversion was required for prion-associated neurotoxicity. Prion-infected astrocytes markedly accelerated neurodegeneration in homologous cocultures compared to infected single neuronal cultures, despite no detectable neurotoxin release. Finally, PrP(Sc) accumulation in neurons led to neuritic damages and cell death, both potentiated by glutamate and reactive oxygen species. Thus, conversion of neuronal PrP(C) rather than PrP(C)-mediated neurotoxic signaling appears as the main culprit in prion-induced neurodegeneration. We suggest that active prion replication in neurons sensitizes them to environmental stress regulated by neighboring cells, including astrocytes.
Collapse
Affiliation(s)
- Sabrina Cronier
- UR892, Virologie et Immunologie Moléculaires, Institut National de Recherche Agronomique, Jouy-en-Josas, France.
| | | | | | | | | | | | | | | |
Collapse
|
109
|
Linden R, Cordeiro Y, Lima LMTR. Allosteric function and dysfunction of the prion protein. Cell Mol Life Sci 2012; 69:1105-24. [PMID: 21984610 PMCID: PMC11114699 DOI: 10.1007/s00018-011-0847-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 09/16/2011] [Accepted: 09/20/2011] [Indexed: 12/30/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs) are neurodegenerative diseases associated with progressive oligo- and multimerization of the prion protein (PrP(C)), its conformational conversion, aggregation and precipitation. We recently proposed that PrP(C) serves as a cell surface scaffold protein for a variety of signaling modules, the effects of which translate into wide-range functional consequences. Here we review evidence for allosteric functions of PrP(C), which constitute a common property of scaffold proteins. The available data suggest that allosteric effects among PrP(C) and its partners are involved in the assembly of multi-component signaling modules at the cell surface, impose upon both physiological and pathological conformational responses of PrP(C), and that allosteric dysfunction of PrP(C) has the potential to entail progressive signal corruption. These properties may be germane both to physiological roles of PrP(C), as well as to the pathogenesis of the TSEs and other degenerative/non-communicable diseases.
Collapse
Affiliation(s)
- Rafael Linden
- Instituto de Biofísica Carlos Chagas Filho, UFRJ, CCS, Cidade Universitária, Rio de Janeiro, Brazil.
| | | | | |
Collapse
|
110
|
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: 0.6] [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
|
111
|
Bate C, Williams A. Neurodegeneration induced by clustering of sialylated glycosylphosphatidylinositols of prion proteins. J Biol Chem 2012; 287:7935-44. [PMID: 22262833 DOI: 10.1074/jbc.m111.275743] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The transmissible spongiform encephalopathies, more commonly known as the prion diseases, are associated with the production and aggregation of disease-related isoforms of the prion protein (PrP(Sc)). The mechanisms by which PrP(Sc) accumulation causes neurodegeneration in these diseases are poorly understood. In cultured neurons, the addition of PrP(Sc) alters cell membranes, increasing cholesterol, activating cytoplasmic phospholipase A(2) (cPLA(2)), and triggering synapse damage. These effects of PrP(Sc) are dependent upon its glycosylphosphatidylinositol (GPI) anchor, suggesting that it is the increased density of GPIs that occurs following the aggregation of PrP(Sc) molecules that triggers neurodegeneration. This hypothesis was supported by observations that cross-linkage of the normal cellular prion protein (PrP(C)) also increased membrane cholesterol, activated cPLA(2), and triggered synapse damage. These effects were not seen after cross-linkage of Thy-1, another GPI-anchored protein, and were dependent on the GPI anchor attached to PrP(C) containing two acyl chains and sialic acid. We propose that the aggregation of PrP(Sc), or the cross-linkage of PrP(C), causes the clustering of sialic acid-containing GPI anchors at high densities, resulting in altered membrane composition, the pathological activation of cPLA(2), and synapse damage.
Collapse
Affiliation(s)
- Clive Bate
- Department of Pathology and Infectious Diseases, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, United Kingdom.
| | | |
Collapse
|
112
|
Klöhn PC, Farmer M, Linehan JM, O'Malley C, Fernandez de Marco M, Taylor W, Farrow M, Khalili-Shirazi A, Brandner S, Collinge J. PrP antibodies do not trigger mouse hippocampal neuron apoptosis. Science 2012; 335:52. [PMID: 22223800 DOI: 10.1126/science.1215579] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Intraperitoneal administration of ICSM18 and 35, monoclonal antibodies against prion protein (PrP), has been shown to significantly delay the onset of prion disease in mice, and humanized versions are candidate therapeutics for prion and Alzheimer's diseases. However, a previous report of severe and widespread apoptosis after intracerebral injection of anti-PrP monoclonal antibodies raised concerns about such therapy and led to an influential model of prion neurotoxicity via cross-linking of cell surface PrP by disease-related PrP aggregates. In extensive studies including ICSM18 and 35, fully humanized ICSM18, and the previously reported proapoptotic antibodies, we found no evidence of apoptosis, thereby questioning this model of prion neurotoxicity.
Collapse
Affiliation(s)
- Peter-Christian Klöhn
- Medical Research Council (MRC) Prion Unit and Department of Neurodegenerative Disease, University College London Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Abstract
Some prion protein mutations create anchorless molecules that cause Gerstmann-Sträussler-Scheinker (GSS) disease. To model GSS, we generated transgenic mice expressing cellular prion protein (PrP(C)) lacking the glycosylphosphatidyl inositol (GPI) anchor, denoted PrP(ΔGPI). Mice overexpressing PrP(ΔGPI) developed a late-onset, spontaneous neurologic dysfunction characterized by widespread amyloid deposition in the brain and the presence of a short protease-resistant PrP fragment similar to those found in GSS patients. In Tg(PrP,ΔGPI) mice, disease onset could be accelerated either by inoculation with brain homogenate prepared from spontaneously ill animals or by coexpression of membrane-anchored, full-length PrP(C). In contrast, coexpression of N-terminally truncated PrP(Δ23-88) did not affect disease progression. Remarkably, disease from ill Tg(PrP,ΔGPI) mice transmitted to mice expressing wild-type PrP(C), indicating the spontaneous generation of prions.
Collapse
|
114
|
Scalabrino G, Mutti E, Veber D, Rodriguez Menendez V, Novembrino C, Calligaro A, Tredici G. The octapeptide repeat PrPCregion and cobalamin-deficient polyneuropathy of the rat. Muscle Nerve 2011; 44:957-67. [DOI: 10.1002/mus.22225] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
115
|
Ishibashi D, Yamanaka H, Mori T, Yamaguchi N, Yamaguchi Y, Nishida N, Sakaguchi S. Antigenic mimicry-mediated anti-prion effects induced by bacterial enzyme succinylarginine dihydrolase in mice. Vaccine 2011; 29:9321-8. [DOI: 10.1016/j.vaccine.2011.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 09/27/2011] [Accepted: 10/05/2011] [Indexed: 11/29/2022]
|
116
|
Fujita K, Yamaguchi Y, Mori T, Muramatsu N, Miyamoto T, Yano M, Miyata H, Ootsuyama A, Sawada M, Matsuda H, Kaji R, Sakaguchi S. Effects of a brain-engraftable microglial cell line expressing anti-prion scFv antibodies on survival times of mice infected with scrapie prions. Cell Mol Neurobiol 2011; 31:999-1008. [PMID: 21516351 PMCID: PMC11498557 DOI: 10.1007/s10571-011-9696-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 02/23/2011] [Indexed: 11/24/2022]
Abstract
We first verified that a single chain Fv fragment against prion protein (anti-PrP scFv) was secreted by HEK293T cells and prevented prion replication in infected cells. We then stably expressed anti-PrP scFv in brain-engraftable murine microglial cells and intracerebrally injected these cells into mice before or after infection with prions. Interestingly, the injection before or at an early time point after infection attenuated the infection marginally but significantly prolonged survival times of the mice. These suggest that the ex vivo gene transfer of anti-PrP scFvs using brain-engraftable cells could be a possible immunotherapeutic approach against prion diseases.
Collapse
Affiliation(s)
- Koji Fujita
- Division of Molecular Neurobiology, The Institute for Enzyme Research, The University of Tokushima, Kuramoto-cho 3-18-15, Tokushima, 770-8503 Japan
- Department of Clinical Neuroscience, The University of Tokushima Graduate School, Tokushima, 770-8503 Japan
| | - Yoshitaka Yamaguchi
- Division of Molecular Neurobiology, The Institute for Enzyme Research, The University of Tokushima, Kuramoto-cho 3-18-15, Tokushima, 770-8503 Japan
| | - Tsuyoshi Mori
- Division of Molecular Neurobiology, The Institute for Enzyme Research, The University of Tokushima, Kuramoto-cho 3-18-15, Tokushima, 770-8503 Japan
| | - Naomi Muramatsu
- Division of Molecular Neurobiology, The Institute for Enzyme Research, The University of Tokushima, Kuramoto-cho 3-18-15, Tokushima, 770-8503 Japan
| | - Takahito Miyamoto
- Division of Molecular Neurobiology, The Institute for Enzyme Research, The University of Tokushima, Kuramoto-cho 3-18-15, Tokushima, 770-8503 Japan
| | - Masashi Yano
- Division of Molecular Neurobiology, The Institute for Enzyme Research, The University of Tokushima, Kuramoto-cho 3-18-15, Tokushima, 770-8503 Japan
| | - Hironori Miyata
- Animal Research Center, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi, Kitakyushu, 807-8555 Japan
| | - Akira Ootsuyama
- Department of Radiation Biology and Health, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi, Kitakyushu, 807-8555 Japan
| | - Makoto Sawada
- Department of Brain Functions, Division of Stress Adaptation and Protection, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601 Japan
| | - Haruo Matsuda
- Department of Molecular and Applied Biosciences, Graduate School of Biosphere Science, Laboratory of Immunology, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528 Japan
| | - Ryuji Kaji
- Department of Clinical Neuroscience, The University of Tokushima Graduate School, Tokushima, 770-8503 Japan
| | - Suehiro Sakaguchi
- Division of Molecular Neurobiology, The Institute for Enzyme Research, The University of Tokushima, Kuramoto-cho 3-18-15, Tokushima, 770-8503 Japan
| |
Collapse
|
117
|
Bate C, Williams A. Amyloid-β-induced synapse damage is mediated via cross-linkage of cellular prion proteins. J Biol Chem 2011; 286:37955-37963. [PMID: 21900234 DOI: 10.1074/jbc.m111.248724] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cellular prion protein (PrP(C)), which is highly expressed at synapses, was identified as a receptor for the amyloid-β (Aβ) oligomers that are associated with dementia in Alzheimer disease. Here, we report that Aβ oligomers secreted by 7PA2 cells caused synapse damage in cultured neurons via a PrP(C)-dependent process. Exogenous PrP(C) added to Prnp knock-out((0/0)) neurons was targeted to synapses and significantly increased Aβ-induced synapse damage. In contrast, the synapse damage induced by a phospholipase A(2)-activating peptide was independent of PrP(C). In Prnp wild-type((+/+)) neurons Aβ oligomers activated synaptic cytoplasmic phospholipase A(2) (cPLA(2)). In these cells, the addition of Aβ oligomers triggered the translocation of cPLA(2) in synapses to cholesterol dense membranes (lipid rafts) where it formed a complex also containing Aβ and PrP(C). In contrast, the addition of Aβ to Prnp((0/0)) neurons did not activate synaptic cPLA(2), which remained in the cytoplasm and was not associated with Aβ. Filtration assays and non-denaturing gels demonstrated that Aβ oligomers cross-link PrP(C). We propose that it is the cross-linkage of PrP(C) by Aβ oligomers that triggers abnormal activation of cPLA(2) and synapse damage. This hypothesis was supported by our observation that monoclonal antibody mediated cross-linkage of PrP(C) also activated synaptic cPLA(2) and caused synapse damage.
Collapse
Affiliation(s)
- Clive Bate
- Department of Pathology and Infectious Diseases, Royal Veterinary College, Hawkshead Lane, North Mymms, Hertfordshire AL9 7TA, United Kingdom.
| | - Alun Williams
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| |
Collapse
|
118
|
Xu Z, Adrover M, Pastore A, Prigent S, Mouthon F, Comoy E, Rezaei H, Deslys JP. Mechanistic insights into cellular alteration of prion by poly-D-lysine: the role of H2H3 domain. FASEB J 2011; 25:3426-35. [PMID: 21697549 DOI: 10.1096/fj.11-187534] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Misfolding of the prion protein (PrP) is the central feature of prion diseases. The conversion of the normal α-helical PrP(C) into a pathological β-enriched PrP(Sc) constitutes an early event in the infectious process. Several hypotheses, involving different regions of the protein, endeavor to delineate the structural mechanism underlying this change of conformation. All current working hypotheses, however, are based on biophysical and modeling studies, the biological relevance of which still needs to be assessed. We have studied the effect of positively charged polymers on the conversion, using polylysine as a model system, and have investigated a possible mechanism of structural stabilization. We have shown that poly-D-lysine removes proteinase K-resistant PrP from prion-infected SN56 neuroblastoma cells without affecting PrP(C). The effect is enantiospecific since the levorotary isomer, poly-L-lysine, has a markedly weaker effect, likely because of its higher susceptibility to degradation. In vitro cross-linking and NMR studies confirm a direct interaction between polylysine and PrP, which mainly maps to the PrP region containing helices 2 and 3 (H2H3). Interaction prevents conformational conversion and protein aggregation. Our results establish a central role of H2H3 in PrP(Sc) amyloidogenesis and replication and provide biological relevance for the pathological misfolding of this domain.
Collapse
Affiliation(s)
- Zhou Xu
- Commissariat à l'Énergie Atomique, Institute of Emerging Diseases and Innovative Therapies, Service d'Étude des Prions et des Infections Atypiques, Fontenay-aux-Roses, France
| | | | | | | | | | | | | | | |
Collapse
|
119
|
Sandberg MK, Al-Doujaily H, Sharps B, Clarke AR, Collinge J. Prion propagation and toxicity in vivo occur in two distinct mechanistic phases. Nature 2011; 470:540-2. [PMID: 21350487 DOI: 10.1038/nature09768] [Citation(s) in RCA: 241] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 12/17/2010] [Indexed: 12/22/2022]
Abstract
Mammalian prions cause fatal neurodegenerative conditions including Creutzfeldt-Jakob disease in humans and scrapie and bovine spongiform encephalopathy in animals. Prion infections are typically associated with remarkably prolonged but highly consistent incubation periods followed by a rapid clinical phase. The relationship between prion propagation, generation of neurotoxic species and clinical onset has remained obscure. Prion incubation periods in experimental animals are known to vary inversely with expression level of cellular prion protein. Here we demonstrate that prion propagation in brain proceeds via two distinct phases: a clinically silent exponential phase not rate-limited by prion protein concentration which rapidly reaches a maximal prion titre, followed by a distinct switch to a plateau phase. The latter determines time to clinical onset in a manner inversely proportional to prion protein concentration. These findings demonstrate an uncoupling of infectivity and toxicity. We suggest that prions themselves are not neurotoxic but catalyse the formation of such species from PrP(C). Production of neurotoxic species is triggered when prion propagation saturates, leading to a switch from autocatalytic production of infectivity (phase 1) to a toxic (phase 2) pathway.
Collapse
Affiliation(s)
- Malin K Sandberg
- MRC Prion Unit and Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | | | | | | | | |
Collapse
|
120
|
Wisniewski T, Goñi F. Immunomodulation for prion and prion-related diseases. Expert Rev Vaccines 2011; 9:1441-52. [PMID: 21105779 DOI: 10.1586/erv.10.131] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Prion diseases are a unique category of illness, affecting both animals and humans, where the underlying pathogenesis is related to a conformational change of a normal self protein called cellular prion protein to a pathological and infectious conformer known as scrapie prion protein (PrP(Sc)). Currently, all prion diseases lack effective treatment and are universally fatal. Past experiences with bovine spongiform encephalopathy and variant Creutzfeldt-Jakob disease mainly in Europe, as well as the current epidemic of chronic wasting disease in North America, have highlighted the need to develop prophylactic and/or therapeutic approaches. In Alzheimer's disease that, like prion disease, is a conformational neurodegenerative disorder, both passive and active immunization has been shown to be highly effective in model animals at preventing disease and cognitive deficits, with emerging data from human trials suggesting that this approach is able to reduce amyloid-related pathology. However, any immunomodulatory approach aimed at a self-antigen has to finely balance an effective humoral immune response with potential autoimmune toxicity. The prion diseases most commonly acquired by infection typically have the alimentary tract as a portal of infectious agent entry. This makes mucosal immunization a potentially attractive method to produce a local immune response that partially or completely prevents prion entry across the gut barrier, while at the same time producing modulated systemic immunity that is unlikely to be associated with toxicity. Our results using an attenuated Salmonella vaccine strain expressing the prion protein showed that mucosal vaccination can protect against prion infection from a peripheral source, suggesting the feasibility of this approach. It is also possible to develop active and/or passive immunomodulatory approaches that more specifically target PrP(Sc) or target the shared pathological conformer found in numerous conformational disorders. Such approaches could have a significant impact on many of the common age-associated dementias.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Department of Psychiatry, Millhauser Laboratories, Room HN419, New York University School of Medicine, 560 First Avenue, New York, NY 10016, USA.
| | | |
Collapse
|
121
|
|
122
|
Škrlj N, Vranac T, Popović M, Čurin Šerbec V, Dolinar M. Specific binding of the pathogenic prion isoform: development and characterization of a humanized single-chain variable antibody fragment. PLoS One 2011; 6:e15783. [PMID: 21283753 PMCID: PMC3024399 DOI: 10.1371/journal.pone.0015783] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 11/24/2010] [Indexed: 11/18/2022] Open
Abstract
Murine monoclonal antibody V5B2 which specifically recognizes the pathogenic form of the prion protein represents a potentially valuable tool in diagnostics or therapy of prion diseases. As murine antibodies elicit immune response in human, only modified forms can be used for therapeutic applications. We humanized a single-chain V5B2 antibody using variable domain resurfacing approach guided by computer modelling. Design based on sequence alignments and computer modelling resulted in a humanized version bearing 13 mutations compared to initial murine scFv. The humanized scFv was expressed in a dedicated bacterial system and purified by metal-affinity chromatography. Unaltered binding affinity to the original antigen was demonstrated by ELISA and maintained binding specificity was proved by Western blotting and immunohistochemistry. Since monoclonal antibodies against prion protein can antagonize prion propagation, humanized scFv specific for the pathogenic form of the prion protein might become a potential therapeutic reagent.
Collapse
Affiliation(s)
- Nives Škrlj
- Biochemistry Chair, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Vranac
- Department for Production of Diagnostic Reagents and Research, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Mara Popović
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vladka Čurin Šerbec
- Biochemistry Chair, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
- Department for Production of Diagnostic Reagents and Research, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Marko Dolinar
- Biochemistry Chair, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|
123
|
Abstract
Prion diseases in humans and animals are characterized by progressive neurodegeneration and the formation of infectious particles called prions. Both features are intimately linked to a conformational transition of the cellular prion protein (PrP(C)) into aberrantly folded conformers with neurotoxic and self-replicating activities. Interestingly, there is increasing evidence that the infectious and neurotoxic properties of PrP conformers are not necessarily coupled. Transgenic mouse models revealed that some PrP mutants interfere with neuronal function in the absence of infectious prions. Vice versa, propagation of prions can occur without causing neurotoxicity. Consequently, it appears plausible that two partially independent pathways exist, one pathway leading to the propagation of infectious prions and another one that mediates neurotoxic signaling. In this review we will summarize current knowledge of neurotoxic PrP conformers and discuss the role of PrP(C) as a mediator of both stress-protective and neurotoxic signaling cascades.
Collapse
|
124
|
Abstract
The prion diseases are a family of rare neurodegenerative disorders that result from the accumulation of a misfolded isoform of the prion protein (PrP), a normal constituent of the neuronal membrane. Five subtypes constitute the known human prion diseases; kuru, Creutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker syndrome (GSS), fatal insomnia (FI), and variant CJD (vCJD). These subtypes are distinguished, in part, by their clinical phenotype, but primarily by their associated brain histopathology. Evidence suggests these phenotypes are defined by differences in the pathogenic conformation of misfolded PrP. Although the vast majority of cases are sporadic, 10% to 15% result from an autosomal dominant mutation of the PrP gene (PRNP). General phenotype-genotype correlations can be made for the major subtypes of CJD, GSS, and FI. This paper will review some of the general background related to prion biology and detail the clinical and pathologic features of the major prion diseases, with a particular focus on the genetic aspects that result in prion disease or modification of its risk or phenotype.
Collapse
Affiliation(s)
- Khalilah Brown
- Center for Comprehensive Care and Research on Memory Disorders, Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
125
|
Prion protein self-interactions: A gateway to novel therapeutic strategies? Vaccine 2010; 28:7810-23. [DOI: 10.1016/j.vaccine.2010.09.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 08/31/2010] [Accepted: 09/03/2010] [Indexed: 11/19/2022]
|
126
|
Tayebi M, David M, Bate C, Jones D, Taylor W, Morton R, Pollard J, Hawke S. Epitope-specific anti-prion antibodies upregulate apolipoprotein E and disrupt membrane cholesterol homeostasis. J Gen Virol 2010; 91:3105-15. [DOI: 10.1099/vir.0.023838-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
127
|
Singh N, Singh A, Das D, Mohan ML. Redox control of prion and disease pathogenesis. Antioxid Redox Signal 2010; 12:1271-94. [PMID: 19803746 PMCID: PMC2864664 DOI: 10.1089/ars.2009.2628] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 09/22/2009] [Accepted: 10/03/2009] [Indexed: 11/12/2022]
Abstract
Imbalance of brain metal homeostasis and associated oxidative stress by redox-active metals like iron and copper is an important trigger of neurotoxicity in several neurodegenerative conditions, including prion disorders. Whereas some reports attribute this to end-stage disease, others provide evidence for specific mechanisms leading to brain metal dyshomeostasis during disease progression. In prion disorders, imbalance of brain-iron homeostasis is observed before end-stage disease and worsens with disease progression, implicating iron-induced oxidative stress in disease pathogenesis. This is an unexpected observation, because the underlying cause of brain pathology in all prion disorders is PrP-scrapie (PrP(Sc)), a beta-sheet-rich conformation of a normal glycoprotein, the prion protein (PrP(C)). Whether brain-iron dyshomeostasis occurs because of gain of toxic function by PrP(Sc) or loss of normal function of PrP(C) remains unclear. In this review, we summarize available evidence suggesting the involvement of oxidative stress in prion-disease pathogenesis. Subsequently, we review the biology of PrP(C) to highlight its possible role in maintaining brain metal homeostasis during health and the contribution of PrP(Sc) in inducing brain metal imbalance with disease progression. Finally, we discuss possible therapeutic avenues directed at restoring brain metal homeostasis and alleviating metal-induced oxidative stress in prion disorders.
Collapse
Affiliation(s)
- Neena Singh
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
128
|
Eiríksson T, Björnsson H, Gudmundsdóttir KB, Kristinsson J, Jóhannesson T. The distribution of four trace elements (Fe, Mn, Cu, Zn) in forage and the relation to scrapie in Iceland. Acta Vet Scand 2010; 52:34. [PMID: 20492671 PMCID: PMC2890668 DOI: 10.1186/1751-0147-52-34] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 05/21/2010] [Indexed: 10/31/2022] Open
Abstract
BACKGROUND Previous studies indicated that the iron (Fe)/manganese (Mn) ratio in forage of sheep was significantly higher on scrapie-afflicted farms than on farms in other scrapie categories. This study was conducted to examine whether Fe and Mn in forage of sheep varied in general according to the scrapie status of different areas in the country. Copper (Cu) and zinc (Zn) were also included because of a possible relation to scrapie. METHODS The country was subdivided into seven Areas (I-VII). Three Areas (I, IV, VII) were designated scrapie-free (never diagnosed or eradicated) and three as scrapie-endemic (II, III, VI); status of Area V was taken as unsettled. Of the harvest 2007 1552 samples were analysed from 344 farms all over the country, mostly grass silage from plastic bales (>90%) and from the first cut (70% or more). Results were expressed as mg kg(-1) dry matter. RESULTS Fe varied enormously from less than 100 mg kg(-1) to 5000 mg kg(-1). Mn varied nearly thirtyfold (17-470 mg kg(-1)). Fe concentration was significantly lower in Area I than in Areas II, V and VI. Mn concentration was significantly higher in Areas I, IV and VII than in Areas II, III, V and VI. The Fe/Mn ratio was significantly less in Area I than in the other areas (except Area IV). Mean Cu concentration was 6.6-8.3 mg kg(-1) and the mean Zn concentration was 24-29 mg kg(-1). They differed significantly in some areas. CONCLUSIONS 1) Fe tended to be in lower amounts in sheep forage in scrapie-free than in endemic areas; 2) Mn was in higher amounts in forage in scrapie-free than endemic areas; 3) the Fe/Mn ratio was lower in scrapie-free than in endemic areas; 4) the Fe/Mn ratio may possibly be used as an indicator of scrapie status; 5) Cu and Zn in sheep forage were not related to scrapie; 6) further study on the role of Fe and Mn in the occurrence of scrapie in Iceland is needed.
Collapse
|
129
|
Shimizu Y, Kaku-Ushiki Y, Iwamaru Y, Muramoto T, Kitamoto T, Yokoyama T, Mohri S, Tagawa Y. A novel anti-prion protein monoclonal antibody and its single-chain fragment variable derivative with ability to inhibit abnormal prion protein accumulation in cultured cells. Microbiol Immunol 2010; 54:112-21. [PMID: 20377745 DOI: 10.1111/j.1348-0421.2009.00190.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
mAbs T1 and T2 were established by immunizing PrP gene ablated mice with recombinant MoPrP of residues 121-231. Both mAbs were cross-reactive with PrP from hamster, sheep, cattle and deer. A linear epitope of mAb T1 was identified at residues 137-143 of MoPrP and buried in PrP(C) expressed on the cell surface. mAb T1 showed no inhibitory effect on accumulation of PrP(Sc) in cultured scrapie-infected neuroblastoma (ScN2a) cells. In contrast, mAb T2 recognized a discontinuous epitope ranged on, or structured by, residues 132-217 and this epitope was exposed on the cell surface PrP(C). mAb T2 showed an excellent inhibitory effect on PrP(Sc) accumulation in vitro at a 50% inhibitory concentration of 0.02 microg/ml (0.14 nM). The scFv form of mAb T2 (scFv T2) was secreted in neuroblastoma (N2a58) cell cultures by transfection through eukaryotic secretion vector. Coculturing of ScN2a cells with scFv T2-producing N2a58 cells induced a clear inhibitory effect on PrP(Sc) accumulation, suggesting that scFv T2 could potentially be an immunotherapeutic tool for prion diseases by inhibition of PrP(Sc) accumulation.
Collapse
Affiliation(s)
- Yoshihisa Shimizu
- Prion Disease Research Center, National Institute of Animal Health, Tsukuba, Ibaraki 305-0856, Japan
| | | | | | | | | | | | | | | |
Collapse
|
130
|
Aguzzi A, O'Connor T. Protein aggregation diseases: pathogenicity and therapeutic perspectives. Nat Rev Drug Discov 2010; 9:237-48. [PMID: 20190788 DOI: 10.1038/nrd3050] [Citation(s) in RCA: 555] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A growing number of diseases seem to be associated with inappropriate deposition of protein aggregates. Some of these diseases--such as Alzheimer's disease and systemic amyloidoses--have been recognized for a long time. However, it is now clear that ordered aggregation of pathogenic proteins does not only occur in the extracellular space, but in the cytoplasm and nucleus as well, indicating that many other diseases may also qualify as amyloidoses. The common structural and pathogenic features of these diverse protein aggregation diseases is only now being fully understood, and may provide novel opportunities for overarching therapeutic approaches such as depleting the monomeric precursor protein, inhibiting aggregation, enhancing aggregate clearance or blocking common aggregation-induced cellular toxicity pathways.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zürich, Schmelzbergstrasse 12, CH8091 Zürich, Switzerland.
| | | |
Collapse
|
131
|
A camelid anti-PrP antibody abrogates PrP replication in prion-permissive neuroblastoma cell lines. PLoS One 2010; 5:e9804. [PMID: 20339552 PMCID: PMC2842437 DOI: 10.1371/journal.pone.0009804] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Accepted: 02/26/2010] [Indexed: 11/19/2022] Open
Abstract
The development of antibodies effective in crossing the blood brain barrier (BBB), capable of accessing the cytosol of affected cells and with higher affinity for PrPSc would be of paramount importance in arresting disease progression in its late stage and treating individuals with prion diseases. Antibody-based therapy appears to be the most promising approach following the exciting report from White and colleagues, establishing the “proof-of-principle” for prion-immunotherapy. After passive transfer, anti-prion antibodies were shown to be very effective in curing peripheral but not central rodent prion disease, due to the fact that these anti-prion antibodies are relatively large molecules and cannot therefore cross the BBB. Here, we show that an anti-prion antibody derived from camel immunised with murine scrapie material adsorbed to immunomagnetic beads is able to prevent infection of susceptible N2a cells and cure chronically scrapie-infected neuroblastoma cultures. This antibody was also shown to transmigrate across the BBB and cross the plasma membrane of neurons to target cytosolic PrPC. In contrast, treatment with a conventional anti-prion antibody derived from mouse immunised with recombinant PrP protein was unable to prevent recurrence of PrPSc replication. Furthermore, our camelid antibody did not display any neurotoxic effects following treatment of susceptible N2a cells as evidenced by TUNEL staining. These findings demonstrate the potential use of anti-prion camelid antibodies for the treatment of prion and other related diseases via non-invasive means.
Collapse
|
132
|
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
|
133
|
Feraudet-Tarisse C, Andreoletti O, Morel N, Simon S, Lacroux C, Mathey J, Lamourette P, Relano A, Torres JM, Creminon C, Grassi J. Immunotherapeutic effect of anti-PrP monoclonal antibodies in transmissible spongiform encephalopathy mouse models: pharmacokinetic and pharmacodynamic analysis. J Gen Virol 2010; 91:1635-45. [DOI: 10.1099/vir.0.018077-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
134
|
Stølsmark T, Tysnes OB. Ny kunnskap om Creutzfeldt-Jakobs sykdom kan gi terapeutiske muligheter. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2010; 130:601-4. [DOI: 10.4045/tidsskr.08.0558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
135
|
Nielsen J, Kulahin N, Walmod PS. Extracellular protein interactions mediated by the neural cell adhesion molecule, NCAM: heterophilic interactions between NCAM and cell adhesion molecules, extracellular matrix proteins, and viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 663:23-53. [PMID: 20017013 DOI: 10.1007/978-1-4419-1170-4_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Janne Nielsen
- Protein Laboratory, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
136
|
Gunther EC, Strittmatter SM. Beta-amyloid oligomers and cellular prion protein in Alzheimer's disease. J Mol Med (Berl) 2009; 88:331-8. [PMID: 19960174 DOI: 10.1007/s00109-009-0568-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 11/13/2009] [Accepted: 11/16/2009] [Indexed: 12/21/2022]
Abstract
Prefibrillar oligomers of the beta-amyloid peptide (A beta) are recognized as potential mediators of Alzheimer's disease (AD) pathophysiology. Deficits in synaptic function, neurotoxicity, and the progression of AD have all been linked to the oligomeric A beta assemblies rather than to A beta monomers or to amyloid plaques. However, the molecular sites of A beta oligomer action have remained largely unknown. Recently, the cellular prion protein (PrP(C)) has been shown to act as a functional receptor for A beta oligomers in brain slices. Because PrP(C) serves as the substrate for Creutzfeldt-Jakob Disease (CJD), these data suggest mechanistic similarities between the two neurodegenerative diseases. Here, we review the importance of A beta oligomers in AD, commonalities between AD and CJD, and the newly emergent role of PrP(C) as a receptor for A beta oligomers.
Collapse
Affiliation(s)
- Erik C Gunther
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 295 Congress Ave., BCMM 436, New Haven, CT 06536-0812, USA
| | | |
Collapse
|
137
|
Massignan T, Stewart RS, Biasini E, Solomon IH, Bonetto V, Chiesa R, Harris DA. A novel, drug-based, cellular assay for the activity of neurotoxic mutants of the prion protein. J Biol Chem 2009; 285:7752-65. [PMID: 19940127 DOI: 10.1074/jbc.m109.064949] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In prion diseases, the infectious isoform of the prion protein (PrP(Sc)) may subvert a normal, physiological activity of the cellular isoform (PrP(C)). A deletion mutant of the prion protein (Delta105-125) that produces a neonatal lethal phenotype when expressed in transgenic mice provides a window into the normal function of PrP(C) and how it can be corrupted to produce neurotoxic effects. We report here the surprising and unexpected observation that cells expressing Delta105-125 PrP and related mutants are hypersensitive to the toxic effects of two classes of antibiotics (aminoglycosides and bleomycin analogues) that are commonly used for selection of stably transfected cell lines. This unusual phenomenon mimics several essential features of Delta105-125 PrP toxicity seen in transgenic mice, including rescue by co-expression of wild type PrP. Cells expressing Delta105-125 PrP are susceptible to drug toxicity within minutes, suggesting that the mutant protein enhances cellular accumulation of these cationic compounds. Our results establish a screenable cellular phenotype for the activity of neurotoxic forms of PrP, and they suggest possible mechanisms by which these molecules could produce their pathological effects in vivo.
Collapse
Affiliation(s)
- Tania Massignan
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | | | | | |
Collapse
|
138
|
Hedlin PD, Cashman NR, Li L, Gupta J, Babiuk LA, Potter AA, Griebel P, Napper S. Design and delivery of a cryptic PrP(C) epitope for induction of PrP(Sc)-specific antibody responses. Vaccine 2009; 28:981-8. [PMID: 19925901 DOI: 10.1016/j.vaccine.2009.10.134] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 10/26/2009] [Accepted: 10/28/2009] [Indexed: 11/30/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs) depend on misfolding of a normal cellular protein (PrP(C)) to an infectious conformation (PrP(Sc)). Targeting PrP(Sc) may represent an effective strategy for immunotherapy while avoiding consequences associated with immune responses to self-proteins. A weakly immunogenic epitope of PrP(C) (YYR), which induces PrP(Sc)-specific antibodies, is used as a starting point for vaccine development. Through optimization of epitope, as well as formulation/delivery, we enhance immunogenicity while retaining PrP(Sc) specificity. In particular, QVYYRPVDQYSNQN, presented by a leukotoxin carrier protein, emerges as a strong vaccine candidate. A vaccine representing this construct induces consistent and sustained serum PrP(Sc)-specific IgG antibody responses following two vaccinations. Antigen specific antibodies are also present within cerebral spinal fluid and mucosal secretions. These characteristics provide a foundation for development of a TSE vaccine.
Collapse
Affiliation(s)
- Peter D Hedlin
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatchewan, Canada
| | | | | | | | | | | | | | | |
Collapse
|
139
|
Sorgato MC, Peggion C, Bertoli A. Is, indeed, the prion protein a Harlequin servant of "many" masters? Prion 2009; 3:202-5. [PMID: 19887913 DOI: 10.4161/pri.3.4.10012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tens of putative interacting partners of the cellular prion protein (PrP(C)) have been identified, yet the physiologic role of PrP(C) remains unclear. For the first time, however, a recent paper has demonstrated that the absence of PrP(C) produces a lethal phenotype. Starting from this evidence, here we discuss the validity of past and more recent literature supporting that, as part of protein platforms at the cell surface, PrP(C) may bridge extracellular matrix molecules and/or membrane proteins to intracellular signaling pathways.
Collapse
Affiliation(s)
- M Catia Sorgato
- Department of Biological Chemistry and CNR Institute of Neuroscience, University of Padova, Padova, Italy
| | | | | |
Collapse
|
140
|
Abstract
Transmissible spongiform encephalopathies (TSEs) are inevitably lethal neurodegenerative diseases that affect humans and a large variety of animals. The infectious agent responsible for TSEs is the prion, an abnormally folded and aggregated protein that propagates itself by imposing its conformation onto the cellular prion protein (PrPC) of the host. PrPCis necessary for prion replication and for prion-induced neurodegeneration, yet the proximal causes of neuronal injury and death are still poorly understood. Prion toxicity may arise from the interference with the normal function of PrPC, and therefore, understanding the physiological role of PrPCmay help to clarify the mechanism underlying prion diseases. Here we discuss the evolution of the prion concept and how prion-like mechanisms may apply to other protein aggregation diseases. We describe the clinical and the pathological features of the prion diseases in human and animals, the events occurring during neuroinvasion, and the possible scenarios underlying brain damage. Finally, we discuss potential antiprion therapies and current developments in the realm of prion diagnostics.
Collapse
|
141
|
Functionally relevant domains of the prion protein identified in vivo. PLoS One 2009; 4:e6707. [PMID: 19738901 PMCID: PMC2733036 DOI: 10.1371/journal.pone.0006707] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Accepted: 07/22/2009] [Indexed: 01/26/2023] Open
Abstract
The prion consists essentially of PrP(Sc), a misfolded and aggregated conformer of the cellular protein PrP(C). Whereas PrP(C) deficient mice are clinically healthy, expression of PrP(C) variants lacking its central domain (PrP(DeltaCD)), or of the PrP-related protein Dpl, induces lethal neurodegenerative syndromes which are repressed by full-length PrP. Here we tested the structural basis of these syndromes by grafting the amino terminus of PrP(C) (residues 1-134), or its central domain (residues 90-134), onto Dpl. Further, we constructed a soluble variant of the neurotoxic PrP(DeltaCD) mutant that lacks its glycosyl phosphatidyl inositol (GPI) membrane anchor. Each of these modifications abrogated the pathogenicity of Dpl and PrP(DeltaCD) in transgenic mice. The PrP-Dpl chimeric molecules, but not anchorless PrP(DeltaCD), ameliorated the disease of mice expressing truncated PrP variants. We conclude that the amino proximal domain of PrP exerts a neurotrophic effect even when grafted onto a distantly related protein, and that GPI-linked membrane anchoring is necessary for both beneficial and deleterious effects of PrP and its variants.
Collapse
|
142
|
Sakaguchi S, Ishibashi D, Matsuda H. Antibody-based immunotherapeutic attempts in experimental animal models of prion diseases. Expert Opin Ther Pat 2009; 19:907-17. [PMID: 19514955 DOI: 10.1517/13543770902988530] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND There has been a dramatic decrease in the risk of transmission of bovine spongiform encephalopathy to humans. In contrast, the risk of human-to-human transmission of variant Creutzfeldt-Jakob disease (vCJD) via medical treatments became potentially high since 4 vCJD cases were reported to be possibly transmitted through blood transfusion in the UK. However, no treatments are yet available for curing prion diseases. OBJECTIVE Conversion of the normal prion protein, PrP(C), to the amyloidogenic PrP, PrP(Sc), plays a pivotal role in the pathogenesis. Recently, certain anti-PrP or anti-37/67-kDa laminin receptor (LRP/LR) antibodies were shown to have the potential to cure chronically infected cells, clearing PrP(Sc) from the cells. This has raised the possibility of antibody based-immunotherapy for prion diseases. This article aims to introduce and discuss the recently published attempts of immunotherapy in prion diseases. METHODS Bibliographic research was carried out using the PubMed database. Patent literature was searched using the UK Intellectual Property Office website. RESULTS/CONCLUSION No satisfying consequences in animals could be detected with anti-PrP antibodies directly infused into the brains of animals by the intraventricular route or by anti-PrP or anti-LRP/LR single chain fragment antibodies directly delivered into the brain by virus vector-mediated gene transfer. This is probably because such delivery systems failed to deliver the antibodies to the neurons relevant for the treatments.
Collapse
Affiliation(s)
- Suehiro Sakaguchi
- The University of Tokushima, The Institute for Enzyme Research, Division of Molecular Neurobiology, 3-18-15 Kuramoto-cho, Tokushima, Japan.
| | | | | |
Collapse
|
143
|
Roostaee A, Côté S, Roucou X. Aggregation and amyloid fibril formation induced by chemical dimerization of recombinant prion protein in physiological-like conditions. J Biol Chem 2009; 284:30907-16. [PMID: 19710507 DOI: 10.1074/jbc.m109.057950] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Prion diseases are caused by the conversion of a cellular protein (PrP(C)) into a misfolded, aggregated isoform (PrP(Res)). Misfolding of recombinant PrP(C) in the absence of PrP(Res) template, cellular factors, denaturing agents, or at neutral pH has not been achieved. A number of studies indicate that dimerization of PrP(C) may be a key step in the aggregation process. In an effort to understand the molecular event that may activate misfolding of PrP(C) in more relevant physiological conditions, we tested if enforced dimerization of PrP(C) may induce a conformational change reminiscent of the conversion of PrP(C) to PrP(Res). We used a well described inducible dimerization strategy whereby a chimeric PrP(C) composed of a modified FK506-binding protein (Fv) fused with PrP(C) and termed Fv-PrP is incubated in the presence of a monomeric FK506 or dimerizing AP20187 ligand. Addition of AP20187 but not FK506 to recombinant Fv-PrP (rFv-PrP) in physiological-like conditions resulted in a rapid conformational change characterized by an increase in beta-sheet structure and simultaneous aggregation of the protein. Aggregates were partially resistant to proteinase K and induced the conversion of soluble rFv-PrP in serial seeding experiments. As judged from thioflavin T binding and electron microscopy, aggregates converted to amyloid fibers. Aggregates were toxic to cultured cells, whereas soluble rFv-PrP and amyloid fibers were harmless. This study strongly supports the proposition that dimerization of PrP(C) is a key pathological primary event in the conversion of PrP(C) and may initiate the pathogenesis of prion diseases.
Collapse
Affiliation(s)
- Alireza Roostaee
- Department of Biochemistry, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | | | | |
Collapse
|
144
|
Mehrpour M, Codogno P. Prion protein: From physiology to cancer biology. Cancer Lett 2009; 290:1-23. [PMID: 19674833 DOI: 10.1016/j.canlet.2009.07.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Revised: 07/10/2009] [Accepted: 07/13/2009] [Indexed: 12/26/2022]
Abstract
Prion protein (PrPc) was originally viewed solely as being involved in prion disease, but now several intriguing lines of evidence have emerged indicating that it plays a fundamental role not only in the nervous system, but also throughout the human body. PrPc is expressed most abundantly in the brain, but has also been detected in other non-neuronal tissues as diverse as lymphoid cells, lung, heart, kidney, gastrointestinal tract, muscle, and mammary glands. Recent data indicate that PrPc may be implicated in biology of glioblastoma, breast cancer, prostate and gastric cancer. Over expression of PrPc is correlated to the acquisition by tumor cells of a phenotype for resistance to cell death induced by TNF alpha and TRAIL or antitumor drugs such as paclitaxel and anthracyclines. PrPc may promote tumorigenesis, proliferation and G1/S transition in gastric cancer cells. This review revisits the physiological functions of PrPc, and its possible implications for cancer biology.
Collapse
|
145
|
Caughey B, Baron GS, Chesebro B, Jeffrey M. Getting a grip on prions: oligomers, amyloids, and pathological membrane interactions. Annu Rev Biochem 2009; 78:177-204. [PMID: 19231987 DOI: 10.1146/annurev.biochem.78.082907.145410] [Citation(s) in RCA: 250] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The prion (infectious protein) concept has evolved with the discovery of new self-propagating protein states in organisms as diverse as mammals and fungi. The infectious agent of the mammalian transmissible spongiform encephalopathies (TSE) has long been considered the prototypical prion, and recent cell-free propagation and biophysical analyses of TSE infectivity have now firmly established its prion credentials. Other disease-associated protein aggregates, such as some amyloids, can also have prion-like characteristics under certain experimental conditions. However, most amyloids appear to lack the natural transmissibility of TSE prions. One feature that distinguishes the latter from the former is the glycophosphatidylinositol membrane anchor on prion protein, the molecule that is corrupted in TSE diseases. The presence of this anchor profoundly affects TSE pathogenesis, which involves major membrane distortions in the brain, and may be a key reason for the greater neurovirulence of TSE prions relative to many other autocatalytic protein aggregates.
Collapse
Affiliation(s)
- Byron Caughey
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA.
| | | | | | | |
Collapse
|
146
|
Single-chain Fv antibody fragments retain binding properties of the monoclonal antibody raised against peptide P1 of the human prion protein. Appl Biochem Biotechnol 2009; 160:1808-21. [PMID: 19597999 DOI: 10.1007/s12010-009-8699-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Accepted: 06/19/2009] [Indexed: 10/20/2022]
Abstract
Prion diseases are incurable neurodegenerative diseases that affect both humans and animals. The infectious agent is a pathogenic form of the prion protein that accumulates in brain as amyloids. Currently, there is neither cure nor reliable preclinical diagnostics on the market available. The growing number of reports shows that passive immunisation is one of the most promising strategies for prion disease therapy, where antibodies against prions may prevent and even cure the infection. Since antibodies are large molecules and, thus, might not be suitable for the therapy, different antibody fragments are a good alternative. Therefore, we have designed and prepared single-chain antibody fragments (scFvs) derived from the PrP(Sc)-specific murine monoclonal antibody V5B2. Using a new expression vector pMD204, we produced scFvs in two opposing chain orientations in the periplasm of Escherichia coli. Both recombinant antibody fragments retained the specificity of the parent antibody and one of these exhibited binding properties comparable to the corresponding murine Fab fragments with the affinity in nM range. Our monovalent antibody fragments are of special interest in view of possible therapeutic reagents for prion diseases as well as for development of a new generation of diagnostics.
Collapse
|
147
|
Nuvolone M, Aguzzi A, Heikenwalder M. Cells and prions: A license to replicate. FEBS Lett 2009; 583:2674-84. [DOI: 10.1016/j.febslet.2009.06.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 06/01/2009] [Accepted: 06/09/2009] [Indexed: 10/20/2022]
|
148
|
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
|
149
|
Colja Venturini A, Bresjanac M, Vranac T, Koren S, Narat M, Popović M, Curin Serbec V. Anti-idiotypic antibodies: a new approach in prion research. BMC Immunol 2009; 10:16. [PMID: 19298674 PMCID: PMC2666643 DOI: 10.1186/1471-2172-10-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Accepted: 03/19/2009] [Indexed: 12/03/2022] Open
Abstract
Background In certain cases, anti-idiotypic antibodies that recognize an antigen-combining site of an antibody can mimic the structure and/or function of certain nominal antigens. This feature makes them particularly useful if conventional experimental approaches fail to fulfil expectations, especially when the molecule of interest is infectious, toxic or difficult to isolate and purify. We suggest the application of an anti-idiotype concept to the field of prion biology, with the aim of evoking a humoral immune response against the pathological isoform of the prion protein (PrPSc). Different ways to induce anti-idiotypic responses were studied in mice and chickens using various forms of V5B2, a PrPSc-specific monoclonal antibody we have described previously. Results The preparation of anti-idiotypic monoclonal antibodies was achieved with well-defined strategies of immunization, selection and subsequent characterization. Our results demonstrate that it is possible to induce a strong anti-idiotypic immune response against the V5B2 monoclonal antibody in both xenogeneic and syngeneic experimental systems. From the competition seen between polyclonal and monoclonal anti-idiotypic antibodies and the original immunogen, the P1 peptide, and even more importantly, the ultimate target antigen, PrPSc, we conclude that selected antibodies bind to the antigen-combining site of the V5B2 monoclonal antibody and might even resemble the PrPSc-specific epitope. The involvement of both antigen-combining sites in the interaction between V5B2 and the most promising monoclonal anti-idiotypic antibody was further supported by molecular docking. Conclusion The results of the present study not only provide an example of the successful production of Ab2 monoclonal antibodies based on a well planned strategy for selection, but should also provide a new experimental approach that is applicable to the field of prion diseases.
Collapse
Affiliation(s)
- Anja Colja Venturini
- Department for Production of Diagnostic Reagents and Research, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia.
| | | | | | | | | | | | | |
Collapse
|
150
|
Abstract
Prions are infectious proteins responsible for a group of fatal neurodegenerative diseases called TSEs (transmissible spongiform encephalopathies) or prion diseases. In mammals, prions reproduce themselves by recruiting the normal cellular protein PrP(C) and inducing its conversion into the disease-causing isoform denominated PrP(Sc). Recently, anti-prion antibodies have been shown to permanently cure prion-infected cells. However, the inability of full-length antibodies and proteins to cross the BBB (blood-brain barrier) hampers their use in the therapy of TSEs in vivo. Alternatively, brain delivery of prion-specific scFv (single-chain variable fragment) by AAV (adeno-associated virus) transfer delays the onset of the disease in infected mice, although protection is not complete. We investigated the anti-prion effects of a recombinant anti-PrP (D18) scFv by direct addition to scrapie-infected cell cultures or by infection with both lentivirus and AAV-transducing vectors. We show that recombinant anti-PrP scFv is able to reduce proteinase K-resistant PrP content in infected cells. In addition, we demonstrate that lentiviruses are more efficient than AAV in gene transfer of the anti-PrP scFv gene and in reducing PrP(Sc) content in infected neuronal cell lines. Finally, we have used a bioinformatic approach to construct a structural model of the D18scFv-PrP(C) complex. Interestingly, according to the docking results, Arg(PrP)(151) (Arg(151) from prion protein) is the key residue for the interactions with D18scFv, anchoring the PrP(C) to the cavity of the antibody. Taken together, these results indicate that combined passive and active immunotherapy targeting PrP might be promising strategies for therapeutic intervention in prion diseases.
Collapse
|