201
|
Ashok A, Hegde RS. Retrotranslocation of prion proteins from the endoplasmic reticulum by preventing GPI signal transamidation. Mol Biol Cell 2008; 19:3463-76. [PMID: 18508914 DOI: 10.1091/mbc.e08-01-0087] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Neurodegeneration in diseases caused by altered metabolism of mammalian prion protein (PrP) can be averted by reducing PrP expression. To identify novel pathways for PrP down-regulation, we analyzed cells that had adapted to the negative selection pressure of stable overexpression of a disease-causing PrP mutant. A mutant cell line was isolated that selectively and quantitatively routes wild-type and various mutant PrPs for ER retrotranslocation and proteasomal degradation. Biochemical analyses of the mutant cells revealed that a defect in glycosylphosphatidylinositol (GPI) anchor synthesis leads to an unprocessed GPI-anchoring signal sequence that directs both ER retention and efficient retrotranslocation of PrP. An unprocessed GPI signal was sufficient to impart ER retention, but not retrotranslocation, to a heterologous protein, revealing an unexpected role for the mature domain in the metabolism of misprocessed GPI-anchored proteins. Our results provide new insights into the quality control pathways for unprocessed GPI-anchored proteins and identify transamidation of the GPI signal sequence as a step in PrP biosynthesis that is absolutely required for its surface expression. As each GPI signal sequence is unique, these results also identify signal recognition by the GPI-transamidase as a potential step for selective small molecule perturbation of PrP expression.
Collapse
Affiliation(s)
- Aarthi Ashok
- Cell Biology and Metabolism Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
202
|
Aguzzi A, Sigurdson C, Heikenwaelder M. Molecular mechanisms of prion pathogenesis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2008; 3:11-40. [PMID: 18233951 DOI: 10.1146/annurev.pathmechdis.3.121806.154326] [Citation(s) in RCA: 264] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Prion diseases are infectious neurodegenerative diseases occurring in humans and animals with an invariably lethal outcome. One fundamental mechanistic event in prion diseases is the aggregation of aberrantly folded prion protein into large amyloid plaques and fibrous structures associated with neurodegeneration. The cellular prion protein (PrPC) is absolutely required for disease development, and prion knockout mice are not susceptible to prion disease. Prions accumulate not only in the central nervous system but also in lymphoid organs, as shown for new variant and sporadic Creutzfeldt-Jakob patients and for some animals. To date it is largely accepted that prions consist primarily of PrPSc, a misfolded and aggregated beta-sheet-rich isoform of PrPC. However, PrPSc may or may not be completely congruent with the infectious moiety. Here, we discuss the molecular mechanisms leading to neurodegeneration, the role of the immune system in prion pathogenesis, and the existence of prion strains that appear to have different tropisms and biochemical characteristics.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zürich, CH-8091 Zürich, Switzerland.
| | | | | |
Collapse
|
203
|
Colombo MC, Vandevondele J, Van Doorslaer S, Laio A, Guidoni L, Rothlisberger U. Copper binding sites in the C-terminal domain of mouse prion protein: A hybrid (QM/MM) molecular dynamics study. Proteins 2008; 70:1084-98. [PMID: 17876822 DOI: 10.1002/prot.21604] [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] [Indexed: 11/10/2022]
Abstract
We present a hybrid QM/MM Car-Parrinello molecular dynamics study of the copper-loaded C-terminal domain of the mouse prion protein. By means of a statistical analysis of copper coordination in known protein structures, we localized the protein regions with the highest propensity for copper ion binding. The identified candidate structures were subsequently refined via QM/MM simulations. Their EPR characteristics were computed to make contact with the experimental data and to probe the sensitivity to structural and chemical changes. Overall best agreement with the experimental EPR data (Van Doorslaer et al., J Phys Chem B 2001; 105: 1631-1639) and the information currently available in the literature is observed for a binding site involving H187. Moreover, a reinterpretation of the experimental proton hyperfine couplings was possible in the light of the present computational findings.
Collapse
Affiliation(s)
- Maria Carola Colombo
- Laboratory of Computational Chemistry and Biochemistry, Institute of Chemical Sciences and Engineering, EPFL, CH-1015 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
204
|
Parkyn CJ, Vermeulen EGM, Mootoosamy RC, Sunyach C, Jacobsen C, Oxvig C, Moestrup S, Liu Q, Bu G, Jen A, Morris RJ. LRP1 controls biosynthetic and endocytic trafficking of neuronal prion protein. J Cell Sci 2008; 121:773-83. [PMID: 18285446 DOI: 10.1242/jcs.021816] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The trafficking of normal cellular prion protein (PrPC) is believed to control its conversion to the altered conformation (designated PrPSc) associated with prion disease. Although anchored to the membrane by means of glycosylphosphatidylinositol (GPI), PrPC on neurons is rapidly and constitutively endocytosed by means of coated pits, a property dependent upon basic amino acids at its N-terminus. Here, we show that low-density lipoprotein receptor-related protein 1 (LRP1), which binds to multiple ligands through basic motifs, associates with PrPC during its endocytosis and is functionally required for this process. Moreover, sustained inhibition of LRP1 levels by siRNA leads to the accumulation of PrPC in biosynthetic compartments, with a concomitant lowering of surface PrPC, suggesting that LRP1 expedites the trafficking of PrPC to the neuronal surface. PrPC and LRP1 can be co-immunoprecipitated from the endoplasmic reticulum in normal neurons. The N-terminal domain of PrPC binds to purified human LRP1 with nanomolar affinity, even in the presence of 1 μM of the LRP-specific chaperone, receptor-associated protein (RAP). Taken together, these data argue that LRP1 controls both the surface, and biosynthetic, trafficking of PrPC in neurons.
Collapse
Affiliation(s)
- Celia J. Parkyn
- King's College London, Wolfson Centre for Age Related Disease, Guy's Campus, London SE1 1UL, UK
| | | | - Roy C. Mootoosamy
- King's College London, Wolfson Centre for Age Related Disease, Guy's Campus, London SE1 1UL, UK
| | - Claire Sunyach
- King's College London, Wolfson Centre for Age Related Disease, Guy's Campus, London SE1 1UL, UK
| | - Christian Jacobsen
- Department of Medical Biochemistry, University of Aarhus, DK-8000 Aarhus C, Denmark
| | - Claus Oxvig
- Department of Medical Biochemistry, University of Aarhus, DK-8000 Aarhus C, Denmark
| | - Søren Moestrup
- Department of Medical Biochemistry, University of Aarhus, DK-8000 Aarhus C, Denmark
| | - Qiang Liu
- Department of Pediatrics, Washington University School of Medicine, St Louis Children's Hospital, St Louis MO 63110, USA
| | - Guojun Bu
- Department of Pediatrics, Washington University School of Medicine, St Louis Children's Hospital, St Louis MO 63110, USA
| | - Angela Jen
- King's College London, Wolfson Centre for Age Related Disease, Guy's Campus, London SE1 1UL, UK
| | - Roger J. Morris
- King's College London, Wolfson Centre for Age Related Disease, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
205
|
Groschup MH, Buschmann A. Rodent models for prion diseases. Vet Res 2008; 39:32. [PMID: 18284909 DOI: 10.1051/vetres:2008008] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2007] [Accepted: 01/15/2008] [Indexed: 11/14/2022] Open
Abstract
Until today most prion strains can only be propagated and the infectivity content assayed by experimentally challenging conventional or transgenic animals. Robust cell culture systems are not available for any of the natural and only for a few of the experimental prion strains. Moreover, the pathogenesis of different transmissible spongiform encephalopathies (TSE) can be analysed systematically by using experimentally infected animals. While, in the beginning, animals belonging to the natural host species were used, more and more rodent model species have been established, mostly due to practical reasons. Nowadays, most of these experiments are performed using highly susceptible transgenic mouse lines expressing cellular prion proteins, PrP, from a variety of species like cattle, sheep, goat, cervidae, elk, hamster, mouse, mink, pig, and man. In addition, transgenic mice carrying specific mutations or polymorphisms have helped to understand the molecular pathomechanisms of prion diseases. Transgenic mouse models have been utilised to investigate the physiological role of PrP(C), molecular aspects of species barrier effects, the cell specificity of the prion propagation, the role of the PrP glycosylation, the mechanisms of the prion spread, the neuropathological roles of PrP(C) and of its abnormal isoform PrP(D) (D for disease) as well as the function of PrP Doppel. Transgenic mouse models have also been used for mapping of PrP regions involved in or required for the PrP conversion and prion replication as well as for modelling of familial forms of human prion diseases.
Collapse
Affiliation(s)
- Martin H Groschup
- Friedrich-Loeffler-Institut , Institute for Novel and Emerging Infectious Diseases, Südufer 10, 17493 Greifswald - Insel Riems, Germany.
| | | |
Collapse
|
206
|
Winklhofer KF, Tatzelt J, Haass C. The two faces of protein misfolding: gain- and loss-of-function in neurodegenerative diseases. EMBO J 2008; 27:336-49. [PMID: 18216876 PMCID: PMC2234348 DOI: 10.1038/sj.emboj.7601930] [Citation(s) in RCA: 291] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Accepted: 10/24/2007] [Indexed: 12/15/2022] Open
Abstract
The etiologies of neurodegenerative diseases may be diverse; however, a common pathological denominator is the formation of aberrant protein conformers and the occurrence of pathognomonic proteinaceous deposits. Different approaches coming from neuropathology, genetics, animal modeling and biophysics have established a crucial role of protein misfolding in the pathogenic process. However, there is an ongoing debate about the nature of the harmful proteinaceous species and how toxic conformers selectively damage neuronal populations. Increasing evidence indicates that soluble oligomers are associated with early pathological alterations, and strikingly, oligomeric assemblies of different disease-associated proteins may share common structural features. A major step towards the understanding of mechanisms implicated in neuronal degeneration is the identification of genes, which are responsible for familial variants of neurodegenerative diseases. Studies based on these disease-associated genes illuminated the two faces of protein misfolding in neurodegeneration: a gain of toxic function and a loss of physiological function, which can even occur in combination. Here, we summarize how these two faces of protein misfolding contribute to the pathomechanisms of Alzheimer's disease, frontotemporal lobar degeneration, Parkinson's disease and prion diseases.
Collapse
Affiliation(s)
- Konstanze F Winklhofer
- Neurobiochemisty, Department of Biochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University, Munich, Germany
| | - Jörg Tatzelt
- Neurobiochemisty, Department of Biochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University, Munich, Germany
| | - Christian Haass
- Center for Integrated Protein Science Munich and Laboratory for Neurodegenerative Disease Research, Department of Biochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
207
|
Sakudo A, Wu G, Onodera T, Ikuta K. Octapeptide repeat region of prion protein (PrP) is required at an early stage for production of abnormal prion protein in PrP-deficient neuronal cell line. Biochem Biophys Res Commun 2008; 365:164-9. [PMID: 17981146 DOI: 10.1016/j.bbrc.2007.10.158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Accepted: 10/25/2007] [Indexed: 11/30/2022]
Abstract
An abnormal isoform of prion protein (PrP(Sc)), which is composed of the same amino acids as cellular PrP (PrP(C)) and has proteinase K (PK)-resistance, hypothetically converts PrP(C) into PrP(Sc). To investigate the region important for PrP(Sc) production, we examined the levels of PrP(Sc) in PrP gene-deficient cells (HpL3-4) expressing PrP(C) deleted of various regions including the octapeptide repeat region (OR) or hydrophobic region (HR). After Chandler or Obihiro prion infection, PrP(Sc) was produced in HpL3-4 cells expressing wild-type PrP(C) or PrP(C) deleted of HR at an early stage and further reduced to below the detectable level, whereas cells expressing PrP(C) deleted of OR showed no PrP(Sc) production. The results suggest that OR of PrP(C) is required for the early step of efficient PrP(Sc) production.
Collapse
Affiliation(s)
- Akikazu Sakudo
- Department of Virology, Center for Infectious Disease Control, Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | |
Collapse
|
208
|
Abstract
Prions represent a new biological paradigm of protein-mediated information transfer. In mammals, prions are the cause of fatal, transmissible neurodegenerative diseases, often referred to as transmissible spongiform encephalopathies. Many unresolved issues remain, including the exact molecular nature of the prion, the detailed mechanism of prion propagation, and the mechanism by which prion diseases can be both genetic and infectious. In addition, we know little about the mechanism by which neurons degenerate during prion diseases. Tied to this, the physiological function of the normal form of the prion protein remains unclear, and it is uncertain whether loss of this function contributes to prion pathogenesis. The factors governing the transmission of prions between species remain unclear, in particular the means by which prion strains and PrP primary structure interact to affect interspecies prion transmission. Despite all these unknowns, dramatic advances in our understanding of prions have occurred because of their transmissibility to experimental animals and the development of transgenic mouse models has done much to further our understanding about various aspects of prion biology. In this chapter, I review recent advances in our understanding of prion biology that derive from this powerful and informative approach.
Collapse
Affiliation(s)
- Glenn C Telling
- Department of Microbiology, Immunology and Molecular Genetics, Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
209
|
Hu W, Kieseier B, Frohman E, Eagar TN, Rosenberg RN, Hartung HP, Stüve O. Prion proteins: Physiological functions and role in neurological disorders. J Neurol Sci 2008; 264:1-8. [PMID: 17707411 DOI: 10.1016/j.jns.2007.06.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2007] [Revised: 05/01/2007] [Accepted: 06/08/2007] [Indexed: 02/01/2023]
Abstract
Stanley Prusiner was the first to promote the concept of misfolded proteins as a cause for neurological disease. It has since been shown by him and other investigators that the scrapie isoform of prion protein (PrP(Sc)) functions as an infectious agent in numerous human and non-human disorders of the central nervous system (CNS). Interestingly, other organ systems appear to be less affected, and do not appear to lead to major co-morbidities. The physiological function of the endogenous cellular form of the prion protein (PrP(C)) is much less clear. It is intriguing that PrP(c) is expressed on most tissues in mammals, suggesting not only biological functions outside the CNS, but also a role other than the propagation of its misfolded isotype. In this review, we summarize accumulating in vitro and in vivo evidence regarding the physiological functions of PrP(C) in the nervous system, as well as in lymphoid organs.
Collapse
Affiliation(s)
- Wei Hu
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, TX 75390-9036, United States
| | | | | | | | | | | | | |
Collapse
|
210
|
Thaa B, Zahn R, Matthey U, Kroneck PMH, Bürkle A, Fritz G. The deletion of amino acids 114-121 in the TM1 domain of mouse prion protein stabilizes its conformation but does not affect the overall structure. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1783:1076-84. [PMID: 18088603 DOI: 10.1016/j.bbamcr.2007.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2007] [Revised: 11/13/2007] [Accepted: 11/14/2007] [Indexed: 11/17/2022]
Abstract
A mutant of mouse prion protein (PrPC) carrying a deletion of residues 114-121 (PrPDelta114-121) has previously been described to lack convertibility into the scrapie-associated isoform of PrP (PrPSc) and to exhibit a dominant-negative effect on the conversion of wild-type PrPC into PrPSc in living cells. Here we report the characterization of recombinantly expressed PrPDelta114-121 by Fourier-transformation infrared spectroscopy (FTIR) and circular dichroism (CD) spectroscopy. The analysis of spectra revealed an increased antiparallel beta-sheet content in the deletion mutant compared to wild-type PrPC. This additional short beta-sheet stabilized the fold of the mutant protein by DeltaDeltaG(0)'=3.4+/-0.3 kJ mol(-1) as shown by chemical unfolding experiments using guanidine hydrochloride. Secondary structure predictions suggest that the additional beta-sheet in PrPDelta114-121 is close to the antiparallel beta-sheet in PrPC. The high-affinity Cu2+-binding site outside the octarepeats, which is located close to the deletion and involves His110 as a ligand, was not affected, as detected by electron paramagnetic resonance (EPR) spectroscopy, suggesting that Cu2+ binding does not contribute to the protection of PrPDelta114-121 from conversion into PrPSc. We propose that the deletion of residues 114-121 stabilizes the mutant protein. This stabilization most likely does not obstruct the interaction of PrPDelta114-121 with PrPSc but represents an energy barrier that blocks the conversion of PrPDelta114-121 into PrPSc.
Collapse
Affiliation(s)
- Bastian Thaa
- Fachbereich Biologie, Mathematisch-Naturwissenschaftliche Sektion, Universität Konstanz, 78457 Konstanz, Germany
| | | | | | | | | | | |
Collapse
|
211
|
Zomosa-Signoret V, Arnaud JD, Fontes P, Alvarez-Martinez MT, Liautard JP. Physiological role of the cellular prion protein. Vet Res 2007; 39:9. [PMID: 18073096 DOI: 10.1051/vetres:2007048] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 09/21/2007] [Indexed: 01/30/2023] Open
Abstract
The prion protein (PrP) plays a key role in the pathogenesis of prion diseases. However, the normal function of the protein remains unclear. The cellular isoform (PrP(C)) 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. Cell biological studies of PrP contribute to our understanding of PrP(C) function. Like other membrane proteins, PrP(C) is post-translationally processed in the endoplasmic reticulum and Golgi on its way to the cell surface after synthesis. Cell surface PrP(C) constitutively cycles between the plasma membrane and early endosomes via a clathrin-dependent mechanism, a pathway consistent with a suggested role for PrP(C) in cellular trafficking of copper ions. Although PrP(-/-) mice have been reported to have only minor alterations in immune function, PrP(C) is up-regulated in T cell activation and may be expressed at higher levels by specialized classes of lymphocytes. Furthermore, antibody cross-linking of surface PrP(C) modulates T cell activation and leads to rearrangements of lipid raft constituents and increased phosphorylation of signaling proteins. These findings appear to indicate an important but, as yet, ill-defined role in T cell function. Recent work has suggested that PrP(C) is required for self-renewal of haematopoietic stem cells. PrP(C) is highly expressed in the central nervous system, and since this is the major site of prion pathology, most interest has focused on defining the role of PrP(C) in neurones. Although PrP(-/-) mice have a grossly normal neurological phenotype, even when neuronal PrP(C) is knocked out postnatally, they do have subtle abnormalities in synaptic transmission, hippocampal morphology, circadian rhythms, and cognition and seizure threshold. Other postulated neuronal roles for PrP(C) include copper-binding, as an anti- and conversely, pro-apoptotic protein, as a signaling molecule, and in supporting neuronal morphology and adhesion. The prion protein may also function as a metal binding protein such as copper, yielding cellular antioxidant capacity suggesting a role in the oxidative stress homeostasis. Finally, recent observations on the role of PrP(C) in long-term memory open a challenging field.
Collapse
|
212
|
Sakaguchi S. Molecular biology of prion protein and its first homologous protein. THE JOURNAL OF MEDICAL INVESTIGATION 2007; 54:211-23. [PMID: 17878669 DOI: 10.2152/jmi.54.211] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Conformational conversion of the normal cellular isoform of prion protein, PrP(C), a glycoprotein anchored to the cell membrane by a glycosylphosphatidylinositol moiety, into the abnormally folded, amyloidogenic prion protein, PrP(Sc), plays a pivotal role in the pathogenesis of prion diseases. It has been suggested that PrP(C) might be functionally disturbed by constitutive conversion to PrP(Sc) due to either the resulting depletion of PrP(C) or the dominant negative effects of PrP(Sc) on PrP(C) or both. Consistent with this, we and others showed that mice devoid of PrP(C) (PrP-/-) spontaneously developed abnormal phenotypes very similar to the neurological abnormalities of prion diseases, supporting the concept that functional loss of PrP(C) might at least be partly involved in the pathogenesis of the diseases. However, no neuronal cell death could be detected in PrP-/- mice, indicating that the functional loss of PrP(C) alone might not be enough to induce neuronal cell death, one of major pathological hallmarks of prion diseases. Interestingly, it was recently shown that the first identified PrP-like protein, termed PrPLP/Doppel (Dpl), is neurotoxic in the absence of PrP(C), causing Purkinje cell degeneration in the cerebellum of mice. Although it is not understood if PrP(Sc) could have a neurotoxic potential similar to PrPLP/Dpl, it is very interesting to speculate that accumulation of PrP(Sc) and the functional disturbance of PrP(C), both of which are caused by constitutive conversion, might be required for the neurodegeneration in prion diseases.
Collapse
Affiliation(s)
- Suehiro Sakaguchi
- Division of Molecular Neurobiology, The Institute for Enzyme Research, The University of Tokushima, Japan
| |
Collapse
|
213
|
Steele AD, Hetz C, Yi CH, Jackson WS, Borkowski AW, Yuan J, Wollmann RH, Lindquist S. Prion pathogenesis is independent of caspase-12. Prion 2007; 1:243-7. [PMID: 19164919 PMCID: PMC2634538 DOI: 10.4161/pri.1.4.5551] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2007] [Accepted: 01/08/2008] [Indexed: 12/22/2022] Open
Abstract
The pathogenic mechanism(s) underlying neurodegenerative diseases associated with protein misfolding is unclear. Several studies have implicated ER stress pathways in neurodegenerative conditions, including prion disease, amyotrophic lateral sclerosis, Alzheimer's disease and many others. The ER stress response and upregulation of ER stress-responsive chaperones is observed in the brains of patients affected with Creutzfeldt-Jacob disease and in mouse models of prion diseases. In particular, the processing of caspase-12, an ER-localized caspase, correlates with neuronal cell death in prion disease. However, the contribution of caspase-12 to neurodegeneration has not been directly addressed in vivo. We confirm that ER stress is induced and that caspase-12 is proteolytically processed in a murine model of infectious prion disease. To address the causality of caspase-12 in mediating infectious prion pathogenesis, we inoculated mice deficient in caspase-12 with prions. The survival, behavior, pathology and accumulation of proteinase K-resistant PrP are indistinguishable between caspase-12 knockout and control mice, suggesting that caspase-12 is not necessary for mediating the neurotoxic effects of prion protein misfolding.
Collapse
Affiliation(s)
- Andrew D Steele
- Whitehead Institute for Biomedical Research; Howard Hughes Medical Institute; Massachusets Institute of Technology; Cambridge, Massachusetts USA
| | - Claudio Hetz
- Department of Cellular and Molecular Biology; Institute of Biomedical Sciences; FONDAP Center for Molecular Studies of the Cell; University of Chile; Santiago, Chile
| | - Caroline H Yi
- Deptartment of Cell Biology; Harvard Medical School; Boston, Massachusetts USA
| | - Walker S Jackson
- Whitehead Institute for Biomedical Research; Howard Hughes Medical Institute; Massachusets Institute of Technology; Cambridge, Massachusetts USA
| | - Andrew W Borkowski
- Whitehead Institute for Biomedical Research; Howard Hughes Medical Institute; Massachusets Institute of Technology; Cambridge, Massachusetts USA
| | - Junying Yuan
- Deptartment of Cell Biology; Harvard Medical School; Boston, Massachusetts USA
| | - Robert H Wollmann
- Department of Pathology; University of Chicago; Chicago, Illinois USA
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research; Howard Hughes Medical Institute; Massachusets Institute of Technology; Cambridge, Massachusetts USA
| |
Collapse
|
214
|
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: 190] [Impact Index Per Article: 10.6] [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
|
215
|
Watts JC, Drisaldi B, Ng V, Yang J, Strome B, Horne P, Sy MS, Yoong L, Young R, Mastrangelo P, Bergeron C, Fraser PE, Carlson GA, Mount HTJ, Schmitt-Ulms G, Westaway D. The CNS glycoprotein Shadoo has PrP(C)-like protective properties and displays reduced levels in prion infections. EMBO J 2007; 26:4038-50. [PMID: 17703189 PMCID: PMC1950727 DOI: 10.1038/sj.emboj.7601830] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2007] [Accepted: 07/24/2007] [Indexed: 12/12/2022] Open
Abstract
The cellular prion protein, PrPC, is neuroprotective in a number of settings and in particular prevents cerebellar degeneration mediated by CNS-expressed Doppel or internally deleted PrP (‘ΔPrP'). This paradigm has facilitated mapping of activity determinants in PrPC and implicated a cryptic PrPC-like protein, ‘π'. Shadoo (Sho) is a hypothetical GPI-anchored protein encoded by the Sprn gene, exhibiting homology and domain organization similar to the N-terminus of PrP. Here we demonstrate Sprn expression and Sho protein in the adult CNS. Sho expression overlaps PrPC, but is low in cerebellar granular neurons (CGNs) containing PrPC and high in PrPC-deficient dendritic processes. In Prnp0/0 CGNs, Sho transgenes were PrPC-like in their ability to counteract neurotoxic effects of either Doppel or ΔPrP. Additionally, prion-infected mice exhibit a dramatic reduction in endogenous Sho protein. Sho is a candidate for π, and since it engenders a PrPC-like neuroprotective activity, compromised neuroprotective activity resulting from reduced levels may exacerbate damage in prion infections. Sho may prove useful in deciphering several unresolved facets of prion biology.
Collapse
Affiliation(s)
- Joel C Watts
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Bettina Drisaldi
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Vivian Ng
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Jing Yang
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Bob Strome
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Patrick Horne
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Larry Yoong
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | | | - Peter Mastrangelo
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Catherine Bergeron
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Paul E Fraser
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | | - Howard T J Mount
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Gerold Schmitt-Ulms
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - David Westaway
- Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Alberta, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Room 116, Environmental Engineering Building, Edmonton, Alberta, Canada T6G 2M8. Tel.: +780 492 9377; Fax: +780 492 9352; E-mail:
| |
Collapse
|
216
|
Sakudo A, Onodera T, Ikuta K. Prion protein gene-deficient cell lines: powerful tools for prion biology. Microbiol Immunol 2007; 51:1-13. [PMID: 17237594 DOI: 10.1111/j.1348-0421.2007.tb03877.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Prion diseases are zoonotic infectious diseases commonly transmissible among animals via prion infections with an accompanying deficiency of cellular prion protein (PrP(C)) and accumulation of an abnormal isoform of prion protein (PrP(Sc)), which are observed in neurons in the event of injury and disease. To understand the role of PrP(C) in the neuron in health and diseases, we have established an immortalized neuronal cell line HpL3-4 from primary hippocampal cells of prion protein (PrP) gene-deficient mice by using a retroviral vector encoding Simian Virus 40 Large T antigen (SV40 LTag). The HpL3-4 cells exhibit cell-type-specific proteins for the neuronal precursor lineage. Recently, this group and other groups have established PrP-deficient cell lines from many kinds of cell types including glia, fibroblasts and neuronal cells, which will have a broad range of applications in prion biology. In this review, we focus on recently obtained information about PrP functions and possible studies on prion infections using the PrPdeficient cell lines.
Collapse
Affiliation(s)
- Akikazu Sakudo
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Japan.
| | | | | |
Collapse
|
217
|
Gains MJ, LeBlanc AC. Canadian Association of Neurosciences Review: prion protein and prion diseases: the good and the bad. Can J Neurol Sci 2007; 34:126-45. [PMID: 17598589 DOI: 10.1017/s0317167100005953] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In the 1700's a strange new disease affecting sheep was recognized in Europe. The disease later became known as "Scrapie" and was the first of a family of similar diseases affecting a number of species that are now known as the Transmissible Spongiform Encephalopathies (TSEs). The appearance of a new disease in humans linked to the consumption of meat products from infected cattle has stimulated widespread public concern and scientific interest in the prion protein and related diseases. Nearly 300 years after the first report, these diseases still merit the descriptor "strange". This family of diseases is characterized by a unique profile of histological changes, can be transmitted as inherited or acquired diseases, as well as apparent sporadic spontaneous generation of the disease. These diseases are believed by many, to be caused by a unique protein only infectious agent. The "prion protein" (PrPC), a term first coined by Stanley Prusiner in 1982 is crucial to the development of these diseases, apparently by acting as a substrate for an abnormal disease associated form. However, aside from being critical to the pathogenesis of the disease, the function of PrPC, which is expressed in all mammals, has defied definitive description. Several roles have been proposed on the basis of in vitro studies, however, thus far, in vivo confirmation has not been forthcoming. The biological features of PrPC also seem to be unusual. Numerous mouse models have been generated in an attempt to understand the pathogenesis of these diseases. This review summarizes the current state of histological features, the etiologic agent, the normal metabolism and the function of the prion protein, as well as the limitations of the mouse models.
Collapse
Affiliation(s)
- Malcolm J Gains
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada
| | | |
Collapse
|
218
|
Abstract
The biological role of the scrapie isoform of prion protein (PrP(Sc)) as an infectious agent in numerous human and non-human disorders of the central nervous system is well established. In contrast, and despite decades of intensive research, the physiological function of the endogenous cellular form of the prion protein (PrP(C)) remains elusive. In mammals, the ubiquitous expression of PrP(C) suggests biological functions other than its pathological role in propagating the accumulation of its misfolded isotype. Other functions that have been attributed to PrP(C) include signal transduction, synaptic transmission and protection against cell death through the apoptotic pathway. More recently, immunoregulatory properties of PrP(C) have been reported. We review accumulating in vitro and in vivo evidence regarding physiological functions of PrP(C).
Collapse
Affiliation(s)
- W Hu
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Dallas, TX 75390-9036, USA
| | | | | |
Collapse
|
219
|
Grasso D, Grasso G, Guantieri V, Impellizzeri G, La Rosa C, Milardi D, Micera G, Osz K, Pappalardo G, Rizzarelli E, Sanna D, Sóvágó I. Environmental effects on a prion's helix II domain: copper(II) and membrane interactions with PrP180-193 and its analogues. Chemistry 2007; 12:537-47. [PMID: 16163753 DOI: 10.1002/chem.200500534] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
An abnormal interaction between copper and the prion protein is believed to play a pivotal role in the pathogenesis of prion diseases. Copper binding has been mainly attributed to the N-terminal domain of the prion protein, but this hypothesis has recently been challenged in some papers which suggest that the C-terminal domain might also compete for metal anchoring. In particular, the segment corresponding to the helix II region of the prion protein, namely PrP180-193, has been shown both to bind copper and to exhibit a copper-enhanced cytotoxicity, as well as to interact with artificial membranes. The present work is aimed at extending these results by choosing the most representative model of this domain and by determining its copper affinity. With this aim, the different role played by the electrostatic properties of the C- and N-termini of PrP180-193 (VNITIKQHTVTTTT) in determining its conformational behaviour, copper coordination and ability to perturb model membranes was investigated. Owing to the low solubility of PrP180-193, its copper affinity was evaluated by using the shorter PrPAc184-188NH2 (IKQHT) analogue as a model. ESI-MS, ESR, UV/Vis, and CD measurements were carried out on the copper(II)/PrPAc184-188NH2 and copper(II)/PrP180-193NH2 systems, and showed that PrPAc184-188NH2 is a reliable model for the metal interaction with the helix II domain. The affinity of copper(II) for the helix II fragment is higher than that for the octarepeat and PrP106-126 peptides. Finally, the different ability of PrP180-193 analogues to perturb the DPPC model membrane was assessed by DSC measurements. The possible biological consequences of these findings are also discussed briefly.
Collapse
Affiliation(s)
- Domenico Grasso
- Università di Catania, Dipartimento di Scienze Chimiche, Viale Andrea Doria 6, 95125 Catania, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
220
|
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: 1.2] [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
|
221
|
Aguzzi A, Heikenwalder M, Polymenidou M. Insights into prion strains and neurotoxicity. Nat Rev Mol Cell Biol 2007; 8:552-61. [PMID: 17585315 DOI: 10.1038/nrm2204] [Citation(s) in RCA: 247] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs) are neurodegenerative diseases that are caused by prions and affect humans and many animal species. It is now widely accepted that the infectious agent that causes TSEs is PrP(Sc), an aggregated moiety of the host-derived membrane glycolipoprotein PrP(C). Although PrP(C) is encoded by the host genome, prions themselves encipher many phenotypic TSE variants, known as prion strains. Prion strains are TSE isolates that, after inoculation into distinct hosts, cause disease with consistent characteristics, such as incubation period, distinct patterns of PrP(Sc) distribution and spongiosis and relative severity of the spongiform changes in the brain. The existence of such strains poses a fascinating challenge to prion research.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zürich, Schmelzbergstrasse 12, CH-8091 Zürich, Switzerland.
| | | | | |
Collapse
|
222
|
Mitteregger G, Vosko M, Krebs B, Xiang W, Kohlmannsperger V, Nölting S, Hamann GF, Kretzschmar HA. The role of the octarepeat region in neuroprotective function of the cellular prion protein. Brain Pathol 2007; 17:174-83. [PMID: 17388948 PMCID: PMC1859984 DOI: 10.1111/j.1750-3639.2007.00061.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Structural alterations of the cellular prion protein (PrPC) seem to be the core of the pathogenesis of prion diseases. However, the physiological function of PrPC remains an enigma. Cell culture experiments have indicated that PrPC and in particular its N‐terminal octarepeat region together with the phosphatidylinositol 3‐kinase (PI3K)/Akt signaling pathways have a fundamental involvement in neuroprotection and oxidative stress reactions. We used wild‐type mice, PrP knockout (Prnp−/−) animals and transgenic mice that lack the octarepeat region (C4/−) and subjected them to controlled ischemia. We identified an increased cleavage and synthesis of PrPC in ischemic brain areas of wild‐type mice compared with sham controls. The infarct size in Prnp−/− animals was increased threefold when compared with wild‐type mice. The infarct size in C4/− animals was identical to Prnp−/− mice, that is, around three times larger than in wild‐type mice. We showed that the PrP in C4/− mice does not functionally rescue the Prnp−/− phenotype; furthermore it is unable to undergo β cleavage, although an increased amount of C1 fragments was found in ischemic brain areas compared with sham controls. We demonstrated that the N‐terminal octarepeat region has a lead function in PrPC physiology and neuroprotection against oxidative stress in vivo.
Collapse
Affiliation(s)
| | - Milan Vosko
- Department of Neurology, Ludwig‐Maximilians University, Munich, Germany
| | | | - Wei Xiang
- Center for Neuropathology and Prion Research
| | | | | | - Gerhard F. Hamann
- Department of Neurology, Ludwig‐Maximilians University, Munich, Germany
| | | |
Collapse
|
223
|
Dong J, Li A, Yamaguchi N, Sakaguchi S, Harris DA. Doppel induces degeneration of cerebellar Purkinje cells independently of Bax. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:599-607. [PMID: 17569776 PMCID: PMC1934519 DOI: 10.2353/ajpath.2007.070262] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Doppel (Dpl) is a prion protein paralog that causes neurodegeneration when expressed ectopically in the brain. To investigate the cellular mechanism underlying this effect, we analyzed Dpl-expressing transgenic mice in which the gene for the proapoptotic protein Bax had been deleted. We found that Bax deletion does not alter either clinical symptoms or Purkinje cell degeneration in Dpl transgenic mice. In addition, we observed that degenerating Purkinje cells in these animals do not display DNA fragmentation or caspase-3 activation. Our results suggest that non-Bax-dependent pathways mediate the toxic effects of Dpl in Purkinje cells, highlighting a possible role for nonapoptotic mechanisms in the death of these neurons.
Collapse
Affiliation(s)
- Jiaxin Dong
- Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Ave., St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
224
|
Abstract
The biochemical nature and the replication of infectious prions have been intensively studied in recent years. Much less is known about the cellular events underlying neuronal dysfunction and cell death. As the cellular function of the normal cellular isoform of prion protein is not exactly known, the impact of gain of toxic function or loss of function, or a combination of both, in prion pathology is still controversial. There is increasing evidence that the normal cellular isoform of the prion protein is a key mediator in prion pathology. Transgenic models were instrumental in dissecting propagation of prions, disease-associated isoforms of prion protein and amyloid production, and induction of neurodegeneration. Four experimental avenues will be discussed here which address scenarios of inappropriate trafficking, folding, or targeting of the prion protein.
Collapse
Affiliation(s)
- Jörg Tatzelt
- Department of Biochemistry, Neurobiochemistry, Ludwig-Maximilians-University Munich, Germany.
| | | |
Collapse
|
225
|
Heitz S, Lutz Y, Rodeau JL, Zanjani H, Gautheron V, Bombarde G, Richard F, Fuchs JP, Vogel MW, Mariani J, Bailly Y. BAX contributes to Doppel-induced apoptosis of prion-protein-deficient Purkinje cells. Dev Neurobiol 2007; 67:670-86. [PMID: 17443816 DOI: 10.1002/dneu.20366] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Research efforts to deduce the function of the prion protein (PrPc) in knock-out mouse mutants have revealed that large deletions in the PrPc genome result in the ectopic neuronal expression of the prion-like protein Doppel (Dpl). In our analysis of one such line of mutant mice, Ngsk Prnp0/0 (NP0/0), we demonstrate that the ectopic expression of Dpl in brain neurons induces significant levels of cerebellar Purkinje cell (PC) death as early as six months after birth. To investigate the involvement of the mitochondrial proapoptotic factor BAX in the Dpl-induced apoptosis of PCs, we have analyzed the progression of PC death in aging NP0/0:Bax-/- double knockout mutants. Quantitative analysis of cell numbers showed that significantly more PCs survived in NP0/0:Bax-/- double mutants than in the NP0/0:Bax+/+ mutants. However, PC numbers were not restored to wildtype levels or to the increased number of PCs observed in Bax-/- mutants. The partial rescue of NP0/0 PCs suggests that the ectopic expression of Dpl induces both BAX-dependent and BAX-independent pathways of cell death. The activation of glial cells that is shown to be associated topographically with Dpl-induced PC death in the NP0/0:Bax+/+ mutants is abolished by the loss of Bax expression in the double mutant mice, suggesting that chronic inflammation is an indirect consequence of Dpl-induced PC death.
Collapse
Affiliation(s)
- S Heitz
- Département Neurotransmission et Sécrétion Neuroendocrine, Institut des Neurosciences Cellulaires et Intégratives (UMR7168-LC2), CNRS/Université Louis Pasteur, IFR 37 des Neurosciences de Strasbourg, and APHP, Hôpital Charles Foix, Ivry/Seine, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Watts JC, Westaway D. The prion protein family: Diversity, rivalry, and dysfunction. Biochim Biophys Acta Mol Basis Dis 2007; 1772:654-72. [PMID: 17562432 DOI: 10.1016/j.bbadis.2007.05.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 04/26/2007] [Accepted: 05/02/2007] [Indexed: 11/24/2022]
Abstract
The prion gene family currently consists of three members: Prnp which encodes PrP(C), the precursor to prion disease associated isoforms such as PrP(Sc); Prnd which encodes Doppel, a testis-specific protein involved in the male reproductive system; and Sprn which encodes the newest PrP-like protein, Shadoo, which is expressed in the CNS. Although the identification of numerous candidate binding partners for PrP(C) has hinted at possible cellular roles, molecular interpretations of PrP(C) activity remain obscure and no widely-accepted view as to PrP(C) function has emerged. Nonetheless, studies into the functional interrelationships of prion proteins have revealed an interesting phenomenon: Doppel is neurotoxic to cerebellar cells in a manner which can be blocked by either PrP(C) or Shadoo. Further examination of this paradigm may help to shed light on two prominent unanswered questions in prion biology: the functional role of PrP(C) and the neurotoxic pathways initiated by PrP(Sc) in prion disease.
Collapse
Affiliation(s)
- Joel C Watts
- Centre for Research in Neurodegenerative Diseases and Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | | |
Collapse
|
227
|
Westergard L, Christensen HM, Harris DA. The cellular prion protein (PrP(C)): its physiological function and role in disease. BIOCHIMICA ET BIOPHYSICA ACTA 2007; 1772:629-44. [PMID: 17451912 PMCID: PMC1986710 DOI: 10.1016/j.bbadis.2007.02.011] [Citation(s) in RCA: 295] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 02/20/2007] [Accepted: 02/22/2007] [Indexed: 12/13/2022]
Abstract
Prion diseases are caused by conversion of a normal cell-surface glycoprotein (PrP(C)) into a conformationally altered isoform (PrP(Sc)) that is infectious in the absence of nucleic acid. Although a great deal has been learned about PrP(Sc) and its role in prion propagation, much less is known about the physiological function of PrP(C). In this review, we will summarize some of the major proposed functions for PrP(C), including protection against apoptotic and oxidative stress, cellular uptake or binding of copper ions, transmembrane signaling, formation and maintenance of synapses, and adhesion to the extracellular matrix. We will also outline how loss or subversion of the cytoprotective or neuronal survival activities of PrP(C) might contribute to the pathogenesis of prion diseases, and how similar mechanisms are probably operative in other neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | - David A. Harris
- Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110
| |
Collapse
|
228
|
Li A, Piccardo P, Barmada SJ, Ghetti B, Harris DA. Prion protein with an octapeptide insertion has impaired neuroprotective activity in transgenic mice. EMBO J 2007; 26:2777-85. [PMID: 17510630 PMCID: PMC1888682 DOI: 10.1038/sj.emboj.7601726] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2007] [Accepted: 04/20/2007] [Indexed: 01/13/2023] Open
Abstract
Familial prion diseases are due to dominantly inherited, germline mutations in the PRNP gene that encodes the prion protein (PrP). The cellular mechanism underlying the pathogenic effect of these mutations remains uncertain. To investigate whether pathogenic mutations impair a normal, physiological activity of PrP, we have crossed Tg(PG14) mice, which express PrP with an octapeptide insertion associated with an inherited prion dementia, with Tg(PrPDelta32-134) mice. Tg(PrPDelta32-134) mice, which express an N-terminally truncated form of PrP, spontaneously develop a neurodegenerative phenotype that is stoichiometrically reversed by coexpression of wild-type PrP. We find that, at equivalent expression levels, PG14 PrP is significantly less efficient than wild-type PrP in suppressing the development of clinical symptoms and neuropathology in Tg(PrPDelta32-134) mice. Thus, our results suggest that some features of the neurological illness associated with inherited PrP mutations may be attributable to a loss of PrP neuroprotective function. This mechanism stands in contrast to the toxic gain-of-function mechanisms that are usually invoked to explain the pathogenesis of dominantly inherited neurodegenerative disorders.
Collapse
Affiliation(s)
- Aimin Li
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Pedro Piccardo
- Division of Neuropathology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, MD, USA
| | - Sami J Barmada
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Bernardino Ghetti
- Division of Neuropathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David A Harris
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO 63110, USA. Tel.: +1 314 362 4690; Fax: +1 314 747 0940. E-mail:
| |
Collapse
|
229
|
Nicolas O, Gavín R, Braun N, Ureña JM, Fontana X, Soriano E, Aguzzi A, del Río JA. Bcl‐2 overexpression delays caspase‐3 activation and rescues cerebellar degeneration in prion‐deficient mice that overexpress amino‐terminally truncated prion. FASEB J 2007; 21:3107-17. [PMID: 17494993 DOI: 10.1096/fj.06-7827com] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Prnp knockout mice that overexpress an amino-truncated form of PrPc (deltaPrP) are ataxic and display cerebellar cell loss and premature death. Studies on the molecular and intracellular events that trigger cell death in these mutants may contribute to elucidate the functions of PrPc and to the design of treatments for prion disease. Here we examined the effects of Bcl-2 overexpression in neurons on the development of the neurological syndrome and cerebellar pathology of deltaPrP. We show that deltaPrP overexpression activates the stress-associated kinases ERK1-2 in reactive astroglia, p38 and the phosphorylation of p53, which leads to the death of cerebellar neurons in mutant mice. We found that the expression of deltaPrP in cell lines expressing very low levels of PrPc strongly induces the activation of apoptotic pathways, thereby leading to caspase-3 activation and cell death, which can be prevented by coexpressing Bcl-2. Finally, we corroborate in vivo that neuronal-directed Bcl-2 overexpression in deltaPrP mice (deltaPrP Bcl-2) markedly reduces caspase-3 activation, glial activation, and neuronal cell death in cerebellum by improving locomotor deficits and life expectancy.
Collapse
Affiliation(s)
- Oriol Nicolas
- Department of Cell Biology, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
230
|
Maas E, Geissen M, Groschup MH, Rost R, Onodera T, Schätzl H, Vorberg IM. Scrapie infection of prion protein-deficient cell line upon ectopic expression of mutant prion proteins. J Biol Chem 2007; 282:18702-10. [PMID: 17468101 DOI: 10.1074/jbc.m701309200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Expression of the cellular prion protein (PrP(C)) is crucial for susceptibility to prions. In vivo, ectopic expression of PrP(C) restores susceptibility to prions and transgenic mice that express heterologous PrP on a PrP knock-out background have been used extensively to study the role of PrP alterations for prion transmission and species barriers. Here we report that prion protein knock-out cells can be rendered permissive to scrapie infection by the ectopic expression of PrP. The system was used to study the influence of sheep PrP-specific residues in mouse PrP on the infection process with mouse adapted scrapie. These studies reveal several critical residues previously not associated with species barriers and demonstrate that amino acid residue alterations at positions known to have an impact on the susceptibility of sheep to sheep scrapie also drastically influence PrP(Sc) formation by mouse-adapted scrapie strain 22L. Furthermore, our data suggest that amino acid polymorphisms located on the outer surfaces of helix 2 and 3 drastically impact conversion efficiency. In conclusion, this system allows for the fast generation of mutant PrP(Sc) that is entirely composed of transgenic PrP and is, thus, ideally suited for testing if artificial PrP molecules can affect prion replication. Transmission of infectivity generated in HpL3-4 cells expressing altered PrP molecules to mice could also help to unravel the potential influence of mutant PrP(Sc) on host cell tropism and strain characteristics in vivo.
Collapse
Affiliation(s)
- Elke Maas
- Institute of Virology, Technical University of Munich, Troger Strasse 30, 81675 Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
231
|
Steele AD, Lindquist S, Aguzzi A. The prion protein knockout mouse: a phenotype under challenge. Prion 2007; 1:83-93. [PMID: 19164918 DOI: 10.4161/pri.1.2.4346] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The key pathogenic event in prion disease involves misfolding and aggregation of the cellular prion protein (PrP). Beyond this fundamental observation, the mechanism by which PrP misfolding in neurons leads to injury and death remains enigmatic. Prion toxicity may come about by perverting the normal function of PrP. If so, understanding the normal function of PrP may help to elucidate the molecular mechansim of prion disease. Ablation of the Prnp gene, which encodes PrP, was instrumental for determining that the continuous production of PrP is essential for replicating prion infectivity. Since the structure of PrP has not provided any hints to its possible function, and there is no obvious phenotype in PrP KO mice, studies of PrP function have often relied on intuition and serendipity. Here, we enumerate the multitude of phenotypes described in PrP deficient mice, many of which manifest themselves only upon physiological challenge. We discuss the pleiotropic phenotypes of PrP deficient mice in relation to the possible normal function of PrP. The critical question remains open: which of these phenotypes are primary effects of PrP deletion and what do they tell us about the function of PrP?
Collapse
Affiliation(s)
- Andrew D Steele
- Whitehead Institute for Biomedical Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.
| | | | | |
Collapse
|
232
|
Huang S, Liang J, Zheng M, Li X, Wang M, Wang P, Vanegas D, Wu D, Chakraborty B, Hays AP, Chen K, Chen SG, Booth S, Cohen M, Gambetti P, Kong Q. Inducible overexpression of wild-type prion protein in the muscles leads to a primary myopathy in transgenic mice. Proc Natl Acad Sci U S A 2007; 104:6800-5. [PMID: 17420473 PMCID: PMC1871865 DOI: 10.1073/pnas.0608885104] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The prion protein (PrP) level in muscle has been reported to be elevated in patients with inclusion-body myositis, polymyositis, dermatomyositis, and neurogenic muscle atrophy, but it is not clear whether the elevated PrP accumulation in the muscles is sufficient to cause muscle diseases. We have generated transgenic mice with muscle-specific expression of PrP under extremely tight regulation by doxycycline, and we have demonstrated that doxycycline-induced overexpression of PrP strictly limited to muscles leads to a myopathy characterized by increased variation of myofiber size, centrally located nuclei, and endomysial fibrosis, in the absence of intracytoplasmic inclusions, rimmed vacuoles, or any evidence of a neurogenic disorder. The PrP-induced myopathy correlates with accumulation of an N-terminal truncated PrP fragment in the muscle, and the muscular PrP displayed consistent mild resistance to protease digestion. Our findings indicate that overexpression of wild-type PrP in skeletal muscles is sufficient to cause a primary myopathy with no signs of peripheral neuropathy, possibly due to accumulation of a cytotoxic truncated form of PrP and/or PrP aggregation.
Collapse
Affiliation(s)
- Shenghai Huang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Jingjing Liang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Mengjie Zheng
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Xinyi Li
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Meiling Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Ping Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Difernando Vanegas
- Department of Cell Biology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Di Wu
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Bikram Chakraborty
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Arthur P. Hays
- Department of Pathology, Columbia University Medical Center, New York, NY 10032
| | - Ken Chen
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Shu G. Chen
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Stephanie Booth
- Division of Host Genetics and Prion Diseases National Microbiology Laboratory, Winnipeg, MB, Canada R3E 3R2; and
| | - Mark Cohen
- **Institute of Pathology, Case Medical Center, Cleveland, OH 44106
| | - Pierluigi Gambetti
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Qingzhong Kong
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
- To whom correspondence should be addressed.
| |
Collapse
|
233
|
Baumann F, Tolnay M, Brabeck C, Pahnke J, Kloz U, Niemann HH, Heikenwalder M, Rülicke T, Bürkle A, Aguzzi A. Lethal recessive myelin toxicity of prion protein lacking its central domain. EMBO J 2007; 26:538-47. [PMID: 17245436 PMCID: PMC1783444 DOI: 10.1038/sj.emboj.7601510] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Accepted: 11/17/2006] [Indexed: 11/08/2022] Open
Abstract
PrP(C)-deficient mice expressing prion protein variants with large amino-proximal deletions (termed PrP(DeltaF)) suffer from neurodegeneration, which is rescued by full-length PrP(C). We now report that expression of PrP(DeltaCD), a PrP variant lacking 40 central residues (94-134), induces a rapidly progressive, lethal phenotype with extensive central and peripheral myelin degeneration. This phenotype was rescued dose-dependently by coexpression of full-length PrP(C) or PrP(C) lacking all octarepeats. Expression of a PrP(C) variant lacking eight residues (114-121) was innocuous in the presence or absence of full-length PrP(C), yet enhanced the toxicity of PrP(DeltaCD) and diminished that of PrP(DeltaF). Therefore, deletion of the entire central domain generates a strong recessive-negative mutant of PrP(C), whereas removal of residues 114-121 creates a partial agonist with context-dependent action. These findings suggest that myelin integrity is maintained by a constitutively active neurotrophic protein complex involving PrP(C), whose effector domain encompasses residues 94-134.
Collapse
Affiliation(s)
- Frank Baumann
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Markus Tolnay
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Christine Brabeck
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Jens Pahnke
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Ulrich Kloz
- Transgenic Core Facility, German Cancer Research Center, Heidelberg, Germany
| | - Hartmut H Niemann
- Strukturbiologie Helmholtz-Zentrum für Infektionsforschung GmbH, Braunschweig, Germany
| | | | - Thomas Rülicke
- Institute of Laboratory Animal Science and Research Center Biomodels Austria University of Veterinary Medicine Vienna, Vienna, Austria
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
- Department of Pathology, Institute of Neuropathology, University Hospital of Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland. Tel.: +41 1 255 2107; Fax: +41 1 255 4402; E-mail:
| |
Collapse
|
234
|
Li A, Barmada SJ, Roth KA, Harris DA. N-terminally deleted forms of the prion protein activate both Bax-dependent and Bax-independent neurotoxic pathways. J Neurosci 2007; 27:852-9. [PMID: 17251426 PMCID: PMC6672905 DOI: 10.1523/jneurosci.4244-06.2007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Transgenic (Tg) mice expressing prion protein (PrP) with a deletion of the flexible, N-terminal tail encompassing residues 32-134 spontaneously develop ataxia, degeneration of cerebellar granule cells, and vacuolation of white matter in the brain and spinal cord, resulting in death by 3 months of age. These abnormalities are completely abrogated by coexpression of wild-type PrP from a single copy of the endogenous Prn-p gene. A similar but much more severe phenotype is seen in transgenic mice expressing PrP deleted for a conserved block of 21 amino acids (residues 105-125) within the N-terminal tail. The latter animals die within 1 week of birth in the absence of endogenous PrP, and fivefold overexpression of wild-type PrP is required to delay death beyond 1 year. To define the cellular pathways mediating the neurotoxicity of PrPdelta32-134 and PrPdelta105-125, we analyzed the effect of genetically deleting the proapoptotic protein Bax in mice expressing these neurotoxic forms of PrP. We find that Bax deletion in Tg(PrPdelta32-134) mice delays the development of clinical illness and slows apoptosis of cerebellar granule cells but has no effect on white matter degeneration. In contrast, Bax deletion has no effect on the clinical or neuropathological phenotype of Tg(delta105-125) mice. Our results indicate that Bax-related pathways mediate the initial neurotoxic actions of PrPdelta32-134 but that neurodegeneration induced by this protein as well as by PrPdelta105-125 also involves Bax-independent pathways.
Collapse
Affiliation(s)
- Aimin Li
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, and
| | - Sami J. Barmada
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, and
| | - Kevin A. Roth
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - David A. Harris
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, and
| |
Collapse
|
235
|
Meade-White K, Race B, Trifilo M, Bossers A, Favara C, Lacasse R, Miller M, Williams E, Oldstone M, Race R, Chesebro B. Resistance to chronic wasting disease in transgenic mice expressing a naturally occurring allelic variant of deer prion protein. J Virol 2007; 81:4533-9. [PMID: 17314157 PMCID: PMC1900179 DOI: 10.1128/jvi.02762-06] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Prion protein (PrP) is a required factor for susceptibility to transmissible spongiform encephalopathy or prion diseases. In transgenic mice, expression of prion protein (PrP) from another species often confers susceptibility to prion disease from that donor species. For example, expression of deer or elk PrP in transgenic mice has induced susceptibility to chronic wasting disease (CWD), the prion disease of cervids. In the current experiments, transgenic mice expressing two naturally occurring allelic variants of deer PrP with either glycine (G) or serine (S) at residue 96 were found to differ in susceptibility to CWD infection. G96 mice were highly susceptible to infection, and disease appeared starting as early as 160 days postinfection. In contrast, S96 mice showed no evidence of disease or generation of disease-associated protease-resistant PrP (PrPres) over a 600-day period. At the time of clinical disease, G96 mice showed typical vacuolar pathology and deposition of PrPres in many brain regions, and in some individuals, extensive neuronal loss and apoptosis were noted in the hippocampus and cerebellum. Extraneural accumulation of PrPres was also noted in spleen and intestinal tissue of clinically ill G96 mice. These results demonstrate the importance of deer PrP polymorphisms in susceptibility to CWD infection. Furthermore, this deer PrP transgenic model is the first to demonstrate extraneural accumulation of PrPres in spleen and intestinal tissue and thus may prove useful in studies of CWD pathogenesis and transmission by oral or other natural routes of infection.
Collapse
Affiliation(s)
- Kimberly Meade-White
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases/NIH, 903 South Fourth Street, Hamilton, MT 59840, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Li A, Christensen HM, Stewart LR, Roth KA, Chiesa R, Harris DA. Neonatal lethality in transgenic mice expressing prion protein with a deletion of residues 105-125. EMBO J 2007; 26:548-58. [PMID: 17245437 PMCID: PMC1783448 DOI: 10.1038/sj.emboj.7601507] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2006] [Accepted: 11/17/2006] [Indexed: 01/21/2023] Open
Abstract
To identify sequence domains important for the neurotoxic and neuroprotective activities of the prion protein (PrP), we have engineered transgenic mice that express a form of murine PrP deleted for a conserved block of 21 amino acids (residues 105-125) in the unstructured, N-terminal tail of the protein. These mice spontaneously developed a severe neurodegenerative illness that was lethal within 1 week of birth in the absence of endogenous PrP. This phenotype was reversed in a dose-dependent fashion by coexpression of wild-type PrP, with five-fold overexpression delaying death beyond 1 year. The phenotype of Tg(PrPDelta105-125) mice is reminiscent of, but much more severe than, those described in mice that express PrP harboring larger deletions of the N-terminus, and in mice that ectopically express Doppel, a PrP paralog, in the CNS. The dramatically increased toxicity of PrPDelta105-125 is most consistent with a model in which this protein has greatly enhanced affinity for a hypothetical receptor that serves to transduce the toxic signal. We speculate that altered binding interactions involving the 105-125 region of PrP may also play a role in generating neurotoxic signals during prion infection.
Collapse
Affiliation(s)
- Aimin Li
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Heather M Christensen
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Leanne R Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| | - Kevin A Roth
- Department of Pathology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Roberto Chiesa
- Dulbecco Telethon Institute (DTI) and Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | - David A Harris
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
237
|
Richt JA, Kasinathan P, Hamir AN, Castilla J, Sathiyaseelan T, Vargas F, Sathiyaseelan J, Wu H, Matsushita H, Koster J, Kato S, Ishida I, Soto C, Robl JM, Kuroiwa Y. Production of cattle lacking prion protein. Nat Biotechnol 2007; 25:132-8. [PMID: 17195841 PMCID: PMC2813193 DOI: 10.1038/nbt1271] [Citation(s) in RCA: 210] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Accepted: 10/27/2006] [Indexed: 01/12/2023]
Abstract
Prion diseases are caused by propagation of misfolded forms of the normal cellular prion protein PrP(C), such as PrP(BSE) in bovine spongiform encephalopathy (BSE) in cattle and PrP(CJD) in Creutzfeldt-Jakob disease (CJD) in humans. Disruption of PrP(C) expression in mice, a species that does not naturally contract prion diseases, results in no apparent developmental abnormalities. However, the impact of ablating PrP(C) function in natural host species of prion diseases is unknown. Here we report the generation and characterization of PrP(C)-deficient cattle produced by a sequential gene-targeting system. At over 20 months of age, the cattle are clinically, physiologically, histopathologically, immunologically and reproductively normal. Brain tissue homogenates are resistant to prion propagation in vitro as assessed by protein misfolding cyclic amplification. PrP(C)-deficient cattle may be a useful model for prion research and could provide industrial bovine products free of prion proteins.
Collapse
Affiliation(s)
- Jürgen A Richt
- National Animal Disease Center, Agriculture Research Services, United States Department of Agriculture, 2300 Dayton Avenue, Ames, Iowa 50010, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Sakudo A, Nakamura I, Ikuta K, Onodera T. Recent Developments in Prion Disease Research: Diagnostic Tools and In Vitro Cell Culture Models. J Vet Med Sci 2007; 69:329-37. [PMID: 17485919 DOI: 10.1292/jvms.69.329] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
After prion infection, an abnormal isoform of prion protein (PrP(Sc)) converts the cellular isoform of prion protein (PrP(C)) into PrP(Sc). PrP(C)-to-PrP(Sc) conversion leads to PrP(Sc) accumulation and PrP(C) deficiency, contributing etiologically to induction of prion diseases. Presently, most of the diagnostic methods for prion diseases are dependent on PrP(Sc) detection. Highly sensitive/accurate specific detection of PrP(Sc) in many different samples is a prerequisite for attempts to develop reliable detection methods. Towards this goal, several methods have recently been developed to facilitate sensitive and precise detection of PrP(Sc), namely, protein misfolding cyclic amplification, conformation-dependent immunoassay, dissociation-enhanced lanthanide fluorescent immunoassay, capillary gel electrophoresis, fluorescence correlation spectroscopy, flow microbead immunoassay, etc. Additionally, functionally relevant prion-susceptible cell culture models that recognize the complexity of the mechanisms of prion infection have also been pursued, not only in relation to diagnosis, but also in relation to prion biology. Prion protein (PrP) gene-deficient neuronal cell lines that can clearly elucidate PrP(C) functions would contribute to understanding of the prion infection mechanism. In this review, we describe the trend in recent development of diagnostic methods and cell culture models for prion diseases and their potential applications in prion biology.
Collapse
Affiliation(s)
- Akikazu Sakudo
- Department of Molecular Immunology, School of Agricultural and Life Sciences, The University of Tokyo, Japan
| | | | | | | |
Collapse
|
239
|
Abstract
AbstractDoppel is a newly recognized prion-like molecule encoded by a novel gene locus, PRND, located on the same chromosomal region of the prion (PRNP) coding gene. Doppel was considered a paralogue and the first member of the prion-gene family, possibly originated through an ancestral gene duplication event. Prion and doppel have different expression patterns, suggesting that the gene products exhibit different biological functions. Actually, doppel is not involved in the aetiology of the Transmissible Spongiform Encephalopathies (TSEs) or “prion diseases” and is highly expressed only within the testicular tissue, suggesting an important physiological role in the process of spermatogenesis. The restricted spatial and temporal expression profile of doppel has suggested its investigation within particular pathological contexts, such as cancers, showing that it might represent a novel and attractive diagnostic molecular marker and that might provide insights into the regulatory pathways of tumor-cell transformation.
Collapse
|
240
|
Sunyach C, Cisse MA, da Costa CA, Vincent B, Checler F. The C-terminal products of cellular prion protein processing, C1 and C2, exert distinct influence on p53-dependent staurosporine-induced caspase-3 activation. J Biol Chem 2006; 282:1956-63. [PMID: 17121821 DOI: 10.1074/jbc.m609663200] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The cellular prion protein (PrP(c)) undergoes various endopro-teolytic attacks within its N-terminal domain, leading to the production of C-terminal fragments (C) tethered to the plasma membrane and soluble N-terminal peptides (N). One of these cleavages occurs at position 110/111, thereby generating C1 and N1 products. We have reported that disintegrins ADAM-10, -9, and -17 participate either directly or indirectly to this proteolytic event. An alternative proteolytic event taking place around residue 90 yields C2 and N2 fragments. The putative function of these proteolytic fragments remained to be established. We have set up two novel human embryonic kidney 293 cell lines stably overexpressing either C1 or C2. We show that C1 potentiates staurosporine-induced caspase-3 activation through a p53-dependent mechanism. Thus, C1 positively controls p53 transcription and mRNA levels and increases p53-like immunoreactivity and activity. C1-induced caspase-3 activation remained unaffected by the blockade of endocytosis in HEK 293 cells and was abolished in p53-deficient fibroblasts. Conversely, overexpression of the C2 fragment did not significantly sensitize HEK 293 cells to apoptotic stimuli and did not modify p53 mRNA levels or activity. Therefore, the nature of the proteolytic cleavage taking place on PrP(c) yielded C-terminal catabolites with distinct function and could be seen as a switch mechanism controlling the function of the PrP(c) in cell survival.
Collapse
Affiliation(s)
- Claire Sunyach
- Institut de Pharmacologie Moleculaire et cellulaire du CNRS, UMR6097, Université Nice-Sophia-Antipolis, Equipe labellisée Fondation pour la Recherche Médicale, 660 route des Lucioles, Sophia-Antipolis, 06560 Valbonne, France
| | | | | | | | | |
Collapse
|
241
|
Galkin AP, Mironova LN, Zhuravleva GA, Inge-Vechtomov SG. Yeast prions, mammalian amyloidoses, and the problem of proteomic networks. RUSS J GENET+ 2006. [DOI: 10.1134/s1022795406110135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
242
|
Pan Y, Zhao L, Liang J, Liu J, Shi Y, Liu N, Zhang G, Jin H, Gao J, Xie H, Wang J, Liu Z, Fan D. Cellular prion protein promotes invasion and metastasis of gastric cancer. FASEB J 2006; 20:1886-8. [PMID: 16877520 DOI: 10.1096/fj.06-6138fje] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cellular prion protein (PrPc) is a glycosylphosphatidylinositol (GPI) -anchored membrane protein that is highly conserved in mammalian species. PrPc has the characteristics of adhesive molecules and is thought to play a role in cell adhesion and membrane signaling. Here we investigated the possible role of PrPc in the process of invasiveness and metastasis in gastric cancers. PrPc was found to be highly expressed in metastatic gastric cancers compared to nonmetastatic ones by immunohistochemical staining. PrPc significantly promoted the adhesive, invasive, and in vivo metastatic abilities of gastric cancer cell lines SGC7901 and MKN45. PrPc also increased promoter activity and the expression of MMP11 by activating phosphorylated ErK1/2 in gastric cancer cells. MEK inhibitor PD98059 and MMP11 antibody (Ab) significantly inhibited in vitro invasive and in vivo metastatic abilities induced by PrPc. N-terminal fragment (amino acid 24-90) was suggested to be an indispensable region for signal transduction and invasion-promoting function of PrPc. Taken together, the present work revealed a novel function of PrPc that the existence of N-terminal region of PrPc could promote the invasive and metastatic abilities of gastric cancer cells at least partially through activation of MEK/ERK pathway and consequent transactivation of MMP11.
Collapse
Affiliation(s)
- Yanglin Pan
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, the Fourth Military Medical University, 17 Changle Western Rd., Xi'an, Shaanxi Province, 710032, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Abstract
Prion protein inhibits Bax activation and Bax-mediated cell death in primary cultures of human neurons and in MCF-7 cells. To determine whether prion protein can protect against Bax-mediated cell death in vivo, wild-type, null and prion over-expressing mice were subjected to Bax-dependent ethanol induced neuronal apoptotic cell death and the brains were immunostained for active caspase-3 as a downstream marker of Bax activation. Bax activation occurs in all ethanol-injected mice independent of their genotype. A higher level of cell death is present in ethanol-injected null mice than in wild-type and prion over-expressing mice. We conclude that prion protein protects some, but not all neurons, against Bax-mediated cell death in this experimental paradigm.
Collapse
Affiliation(s)
- Malcolm J Gains
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
244
|
Mazzoni IE, Ledebur HC, Paramithiotis E, Cashman N. Lymphoid signal transduction mechanisms linked to cellular prion protein. Biochem Cell Biol 2006; 83:644-53. [PMID: 16234853 DOI: 10.1139/o05-058] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The normal cellular isoform of the prion protein (PrPC) is a glycosylphosphatidylinositol-anchored cell surface protein that is expressed widely, including in lymphoid cells. We compared lectin-induced mitogenesis and selected cell signaling pathways in splenocytes from wild-type BALB/c mice and Zrch Prnp0/0 (PrP0/0) mice bred on a BALB/c background for more than 10 generations. 3H-thymidine incorporation induced by concanavalin A (Con A) or phytohemagglutinin (PHA) was significantly reduced in PrP0/0 splenocytes, most prominently early in activation (24 and 48 h). Con A activation in PrP0/0 splenocytes was associated with differences in the phosphorylation (P) patterns of protein kinase C (PKC alpha/beta, but not delta) and the PKC downstream effectors p44/42MAPK (mitogen-activated protein kinase). P-PKC and P-MAPK profiles were similar in wild-type and PrP0/0 splenocytes following PMA treatment, indicating that the ability of these 2 enzymes to be phosphorylated is not impaired in the absence of PrPC. Con A-induced calcium fluxes, monitored by indo-1 fluorescence, were equivalent in PrP0/0 and PrP+/+ splenocytes, suggesting that calcium-dependent mechanisms are not directly implicated in the differential phosphorylation patterns or mitotic responses. Our data indicate that PrP0/0 splenocytes display defects in upstream or downstream mechanism(s) that modulate PKCalpha/beta phosphorylation, which in turn affects its capacity to regulate splenocyte mitosis, consistent with a role for PrPC in immune function.
Collapse
Affiliation(s)
- I E Mazzoni
- Caprion Pharmaceuticals, Montreal, QC H4S 2C8, Canada
| | | | | | | |
Collapse
|
245
|
Zawlik I, Witusik M, Hulas-Bigoszewska K, Piaskowski S, Szybka M, Golanska E, Liberski PP, Rieske P. Regulation of PrPC expression: Nerve growth factor (NGF) activates the prion gene promoter through the MEK1 pathway in PC12 cells. Neurosci Lett 2006; 400:58-62. [PMID: 16520000 DOI: 10.1016/j.neulet.2006.02.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Revised: 01/12/2006] [Accepted: 02/05/2006] [Indexed: 10/24/2022]
Abstract
A high expression of PrP(C) in cells is one factor that increases the risk of conversion to the misfolded, disease-associated form (PrP(Sc)) characteristic of transmissible spongiform encephalopathies. Thus, developing a method to control the level of PrP(C) expression in cells could be one way to delay or prevent the onset of clinical signs of these diseases. In this study the mechanisms controlling the expression of the Prnp gene in PC12 cells and in rat brain were examined. We observed a slight activation of a cloned fragment of the human PRNP gene promoter using the luciferase reporter system in PC12 cells stimulated with nerve growth factor (NGF). The activating effect of NGF was enhanced by mitogen-activated protein kinase (MEK1) and suppressed by myristylated serine/threonine kinase (myrAKT). These results suggest that MEK1 is a positive activator of the PRNP promoter that inhibits the AKT pathway. Independent experiments suggested that high expression of PrP(C) in the brain depends on the rate of translation and/or the efficiency of PrP(C) stabilization. We also investigated the epigenic status of the Prnp promoter. We observed no increase of PrP(C) or Prnp mRNA levels in PC12 cells after treatment with the DNA-demethylating agent. The Prnp promoter did not display methylation either in NGF-treated and untreated PC12 cells, or in the rat brain. These results improve the understanding of the regulation of the Prnp gene promoter, a DNA regulatory element controlling the expression of PrP(C), a protein involved in several neurological diseases.
Collapse
Affiliation(s)
- Izabela Zawlik
- Department of Molecular Pathology and Neuropathology, Chair of Oncology, Medical University of Lodz, 8/10 Czechoslowacka str., 92-216 Lodz, Poland
| | | | | | | | | | | | | | | |
Collapse
|
246
|
Raeber AJ, Brandner S, Klein MA, Benninger Y, Musahl C, Frigg R, Roeckl C, Fischer MB, Weissmann C, Aguzzi A. Transgenic and knockout mice in research on prion diseases. Brain Pathol 2006; 8:715-33. [PMID: 9804380 PMCID: PMC8098451 DOI: 10.1111/j.1750-3639.1998.tb00197.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Since the discovery of the prion protein (PrP) gene more than a decade ago, transgenetic investigations on the PrP gene have shaped the field of prion biology in an unprecedented way. Many questions regarding the role of PrP in susceptibility of an organism exposed to prions have been elucidated. For example mice with a targeted disruption of the PrP gene have allowed the demonstration that an organism that lacks PrPc is resistant to infection by prions. Reconstitution of these mice with mutant PrP genes allowed investigations on the structure-activity relationship of the PrP gene with regard to scrapie susceptibility. Unexpectedly, transgenic mice expressing PrP with specific amino-proximal truncations spontaneously develop a neurologic syndrome presenting with ataxia and cerebellar lesions. A distinct spontaneous neurologic phenotype was observed in mice with internal deletions in PrP. Using ectopic expression of PrP in PrP knockout mice has turned out to be a valuable approach towards the identification of host cells that are capable of replicating prions. Transgenic mice have also contributed to our understanding of the molecular basis of the species barrier for prions. Finally, the availability of PrP knockout mice and transgenic mice overexpressing PrP allows selective reconstitution experiments aimed at expressing PrP in neurografts or in specific populations of hemato- and lymphopoietic cells. Such studies have shed new light onto the mechanisms of prion spread and disease pathogenesis.
Collapse
Affiliation(s)
- A J Raeber
- Institute of Neuropathology, Department of Pathology, University Hospital, Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Stewart RS, Piccardo P, Ghetti B, Harris DA. Neurodegenerative illness in transgenic mice expressing a transmembrane form of the prion protein. J Neurosci 2006; 25:3469-77. [PMID: 15800202 PMCID: PMC6724892 DOI: 10.1523/jneurosci.0105-05.2005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Although PrP(Sc) is thought to be the infectious form of the prion protein, it may not be the form that is responsible for neuronal cell death in prion diseases. (Ctm)PrP is a transmembrane version of the prion protein that has been proposed to be a neurotoxic intermediate underlying prion-induced pathogenesis. To investigate this hypothesis, we have constructed transgenic mice that express L9R-3AV PrP, a mutant prion protein that is synthesized exclusively in the (Ctm)PrP form in transfected cells. These mice develop a fatal neurological illness characterized by ataxia and marked neuronal loss in the cerebellum and hippocampus. (Ctm)PrP in neurons cultured from transgenic mice is localized to the Golgi apparatus, rather than to the endoplasmic reticulum as in transfected cell lines. Surprisingly, development of the neurodegenerative phenotype is strongly dependent on coexpression of endogenous, wild-type PrP. Our results provide new insights into the cell biology of (Ctm)PrP, the mechanism by which it induces neurodegeneration, and possible cellular activities of PrP(C).
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Arginine/genetics
- Blotting, Western/methods
- Brain/metabolism
- Brain/pathology
- Cells, Cultured
- Cerebellum/cytology
- Cricetinae
- Cricetulus
- Detergents/pharmacology
- Disease Models, Animal
- Electrophoresis, Polyacrylamide Gel/methods
- Fluorescent Antibody Technique/methods
- Gene Expression
- Glial Fibrillary Acidic Protein/metabolism
- Golgi Apparatus/metabolism
- Golgi Matrix Proteins
- Immunoprecipitation/methods
- Leucine/genetics
- Membrane Proteins/metabolism
- Methionine/pharmacokinetics
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutation
- Neurodegenerative Diseases/genetics
- Neurodegenerative Diseases/metabolism
- Neurodegenerative Diseases/physiopathology
- Neurons
- Octoxynol/pharmacology
- PrPSc Proteins/genetics
- PrPSc Proteins/metabolism
- Prion Diseases/genetics
- Prion Diseases/metabolism
- Prion Diseases/physiopathology
- Protein Structure, Tertiary/genetics
- Protein Structure, Tertiary/physiology
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Sulfur Isotopes/pharmacokinetics
- Time Factors
- Type C Phospholipases/pharmacology
- Valine/genetics
Collapse
Affiliation(s)
- Richard S Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
248
|
Radovanovic I, Braun N, Giger OT, Mertz K, Miele G, Prinz M, Navarro B, Aguzzi A. Truncated prion protein and Doppel are myelinotoxic in the absence of oligodendrocytic PrPC. J Neurosci 2006; 25:4879-88. [PMID: 15888663 PMCID: PMC6724775 DOI: 10.1523/jneurosci.0328-05.2005] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The cellular prion protein PrP(C) confers susceptibility to transmissible spongiform encephalopathies, yet its normal function is unknown. Although PrP(C)-deficient mice develop and live normally, expression of amino proximally truncated PrP(C) (DeltaPrP) or of its structural homolog Doppel (Dpl) causes cerebellar degeneration that is prevented by coexpression of full-length PrP(C). We now report that mice expressing DeltaPrP or Dpl suffer from widespread leukoencephalopathy. Oligodendrocyte-specific expression of full-length PrP(C) under control of the myelin basic protein (MBP) promoter repressed leukoencephalopathy and vastly extended survival but did not prevent cerebellar granule cell (CGC) degeneration. Conversely, neuron-specific PrP(C) expression under control of the neuron-specific enolase (NSE) promoter antagonized CGC degeneration but not leukoencephalopathy. PrP(C) was found in purified myelin and in cultured oligodendrocytes of both wild-type and MBP-PrP transgenic mice but not in NSE-PrP mice. These results identify white-matter damage as an extraneuronal PrP-associated pathology and suggest a previously unrecognized role of PrP(C) in myelin maintenance.
Collapse
Affiliation(s)
- Ivan Radovanovic
- Institute of Neuropathology, University Hospital of Zurich, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
249
|
Giese A, Buchholz M, Herms J, Kretzschmar HA. Mouse brain synaptosomes accumulate copper-67 efficiently by two distinct processes independent of cellular prion protein. J Mol Neurosci 2006; 27:347-54. [PMID: 16280605 DOI: 10.1385/jmn:27:3:347] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Accepted: 06/09/2005] [Indexed: 01/24/2023]
Abstract
The prion protein (PrPC) is a copper-binding, cell-surface protein that plays an essential role in the etiology of transmissible spongiform encephalopathies. Atomic absorption spectroscopy studies have established that synaptosomal copper content is reduced in PrPC-deficient mice as compared with wild-type (WT) or PrP- overexpressing mice. To address the question of whether this is the result of a loss of function of PrPC in copper transport across the plasma membrane at the synapse, we have used synaptosomes incubated with 67Cu as a model system to characterize the mechanism of copper accumulation in nerve terminals. Our results demonstrate that mouse brain synaptosomes accumulate copper efficiently by at least two distinct mechanisms. In the presence of 1 mM EDTA, copper was taken up via a saturable high-affinity process that was moderately susceptible to competition by high concentrations of NiCl2. Uptake characteristics were clearly different in the presence of 400 microM histidine, with the most noticeable dissimilarities being considerably elevated uptake rates and moderate competition by ZnCl2 rather than NiCl2. No significant differences in copper uptake capability between WT and PrPC-knockout synaptosomes were observed under any of the experimental conditions tested in this study. Furthermore, preincubation of synaptosomes with an antibody binding to the copper-binding repeat region of the prion protein had no effect on copper uptake either. Taken together, our data indicate that synaptosomal copper uptake is independent of PrPC.
Collapse
Affiliation(s)
- Armin Giese
- Zentrum für Neuropathologie und Prionforschung (ZNP), Ludwig-Maximilians-Universität München, München, Germany.
| | | | | | | |
Collapse
|
250
|
Krebs B, Dorner-Ciossek C, Schmalzbauer R, Vassallo N, Herms J, Kretzschmar HA. Prion protein induced signaling cascades in monocytes. Biochem Biophys Res Commun 2006; 340:13-22. [PMID: 16343423 DOI: 10.1016/j.bbrc.2005.11.158] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2005] [Accepted: 11/25/2005] [Indexed: 11/19/2022]
Abstract
Prion proteins play a central role in transmission and pathogenesis of transmissible spongiform encephalopathies. The cellular prion protein (PrP(C)), whose physiological function remains elusive, is anchored to the surface of a variety of cell types including neurons and cells of the lymphoreticular system. In this study, we investigated the response of a mouse monocyte/macrophage cell line to exposure with PrP(C) fusion proteins synthesized with a human Fc-tag. PrP(C) fusion proteins showed an attachment to the surface of monocyte/macrophages in nanomolar concentrations. This was accompanied by an increase of cellular tyrosine phosphorylation as a result of activated signaling pathways. Detailed investigations exhibited activation of downstream pathways through a stimulation with PrP fusion proteins, which include phosphorylation of ERK(1,2) and Akt kinase. Macrophages opsonize and present antigenic structures, contact lymphocytes, and deliver cytokines. The findings reported here may become the basis of understanding the molecular function of PrP(C) in monocytes and macrophages.
Collapse
Affiliation(s)
- Bjarne Krebs
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, München, Germany
| | | | | | | | | | | |
Collapse
|