151
|
Sakudo A, Onodera T. Prion protein (PrP) gene-knockout cell lines: insight into functions of the PrP. Front Cell Dev Biol 2015; 2:75. [PMID: 25642423 PMCID: PMC4295555 DOI: 10.3389/fcell.2014.00075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/22/2014] [Indexed: 11/13/2022] Open
Abstract
Elucidation of prion protein (PrP) functions is crucial to fully understand prion diseases. A major approach to studying PrP functions is the use of PrP gene-knockout (Prnp (-/-)) mice. So far, six types of Prnp (-/-) mice have been generated, demonstrating the promiscuous functions of PrP. Recently, other PrP family members, such as Doppel and Shadoo, have been found. However, information obtained from comparative studies of structural and functional analyses of these PrP family proteins do not fully reveal PrP functions. Recently, varieties of Prnp (-/-) cell lines established from Prnp (-/-) mice have contributed to the analysis of PrP functions. In this mini-review, we focus on Prnp (-/-) cell lines and summarize currently available Prnp (-/-) cell lines and their characterizations. In addition, we introduce the recent advances in the methodology of cell line generation with knockout or knockdown of the PrP gene. We also discuss how these cell lines have provided valuable insights into PrP functions and show future perspectives.
Collapse
Affiliation(s)
- Akikazu Sakudo
- Laboratory of Biometabolic Chemistry, Faculty of Medicine, School of Health Sciences, University of the Ryukyus Nishihara, Japan
| | - Takashi Onodera
- Research Center for Food Safety, School of Agricultural and Life Sciences, University of Tokyo Tokyo, Japan
| |
Collapse
|
152
|
Sorce S, Nuvolone M, Keller A, Falsig J, Varol A, Schwarz P, Bieri M, Budka H, Aguzzi A. The role of the NADPH oxidase NOX2 in prion pathogenesis. PLoS Pathog 2014; 10:e1004531. [PMID: 25502554 PMCID: PMC4263757 DOI: 10.1371/journal.ppat.1004531] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/20/2014] [Indexed: 11/26/2022] Open
Abstract
Prion infections cause neurodegeneration, which often goes along with oxidative stress. However, the cellular source of reactive oxygen species (ROS) and their pathogenetic significance are unclear. Here we analyzed the contribution of NOX2, a prominent NADPH oxidase, to prion diseases. We found that NOX2 is markedly upregulated in microglia within affected brain regions of patients with Creutzfeldt-Jakob disease (CJD). Similarly, NOX2 expression was upregulated in prion-inoculated mouse brains and in murine cerebellar organotypic cultured slices (COCS). We then removed microglia from COCS using a ganciclovir-dependent lineage ablation strategy. NOX2 became undetectable in ganciclovir-treated COCS, confirming its microglial origin. Upon challenge with prions, NOX2-deficient mice showed delayed onset of motor deficits and a modest, but significant prolongation of survival. Dihydroethidium assays demonstrated a conspicuous ROS burst at the terminal stage of disease in wild-type mice, but not in NOX2-ablated mice. Interestingly, the improved motor performance in NOX2 deficient mice was already measurable at earlier stages of the disease, between 13 and 16 weeks post-inoculation. We conclude that NOX2 is a major source of ROS in prion diseases and can affect prion pathogenesis. The deposition of misfolded, aggregated prion protein in the brain causes transmissible spongiform encephalopathies (TSE), a group of disorders including Creutzfeldt–Jakob disease and mad cow disease. TSE are characterized by neurodegeneration and progressive, lethal neurological dysfunction. Signs of oxidative damage are found in TSE, implying excessive production of reactive oxygen species (ROS), yet their source is unclear. Here, we analyzed the role of the NADPH oxidase enzyme, NOX2, in prion pathogenesis. NOX2 is a membrane-bound electrochemical pump that generates ROS. We found that NOX2 is upregulated in the brains of patients with Creutzfeldt-Jakob disease and of prion-infected mice. Interestingly, NOX2 ablation led to abrogation of ROS production in mice inoculated with prions, and was associated with a milder clinical course of the disease and increased life expectancy. We conclude that NOX2 is a relevant contributor to the excessive production of ROS. This study spawns the possibility that inhibiting NOX2 activation might help attenuate prion disease progression – a legitimate and important goal even if there is little reason to expect anti-NOX2 therapies to be curative.
Collapse
Affiliation(s)
- Silvia Sorce
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Mario Nuvolone
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Annika Keller
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Jeppe Falsig
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Ahmet Varol
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Petra Schwarz
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Monika Bieri
- Institute of Surgical Pathology, University Hospital of Zurich, Zurich, Switzerland
| | - Herbert Budka
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
153
|
Prion-induced and spontaneous formation of transmissible toxicity in PrP transgenic Drosophila. Biochem J 2014; 463:31-40. [PMID: 25000212 DOI: 10.1042/bj20140129] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Prion diseases are fatal transmissible neurodegenerative diseases of various mammalian species. Central to these conditions is the conversion of the normal host prion protein PrP(C) into the abnormal prion conformer PrP(Sc). Mature PrP(C) is attached to the plasma membrane by a glycosylphosphatidylinositol anchor, whereas during biosynthesis and metabolism cytosolic and secreted forms of the protein may arise. The role of topological PrP(C) variants in the mechanism of prion formation and prion-induced neurotoxicity during prion disease remains undefined. In the present study we investigated whether Drosophila transgenic for ovine PrP targeted to the plasma membrane, to the cytosol or for secretion, could produce transmissible toxicity following exposure to exogenous ovine prions. Although all three topological variants of PrP were efficiently expressed in Drosophila, cytosolic PrP was conformationally distinct and required denaturation before recognition by immunobiochemical methods. Adult Drosophila transgenic for pan neuronally expressed ovine PrP targeted to the plasma membrane, to the cytosol or for secretion exhibited a decreased locomotor activity after exposure at the larval stage to ovine prions. Proteinase K-resistant PrP(Sc) was detected by protein misfolding cyclic amplification in prion-exposed Drosophila transgenic for membrane-targeted PrP. Significantly, head homogenate from all three variants of prion-exposed PrP transgenic Drosophila induced a decreased locomotor activity when transmitted to PrP recipient flies. Drosophila transgenic for PrP targeted for secretion exhibited a spontaneous locomotor defect in the absence of prion exposure that was transmissible in PrP transgenic flies. Our data are consistent with the formation of transmissible prions in PrP transgenic Drosophila.
Collapse
|
154
|
Roucou X. Regulation of PrP(C) signaling and processing by dimerization. Front Cell Dev Biol 2014; 2:57. [PMID: 25364762 PMCID: PMC4207009 DOI: 10.3389/fcell.2014.00057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 09/19/2014] [Indexed: 12/15/2022] Open
Abstract
The cellular prion protein (PrPC) is a glycosylphosphatidylinositol (GPI)-anchored protein present at the cell surface. PrPC N-terminal moiety is intrinsically disordered and is able to interact with a variety of ligands. Physiological ligands have neurotrophic activity, whilst others, including protein toxic oligomers, have neurotoxic functions. These two opposite activities involve different interacting partners and result from different PrPC-activated signaling pathways. Remarkably, PrPC may be inactivated either by physiological endoproteolysis and release of the N-terminal domain, or by ectodomain shedding. Ligand-induced PrPC dimerization or enforced dimerization of PrPC indicate that PrPC dimerization represents an important molecular switch for both intracellular signaling and inactivation by the release of PrPC N-terminal domain or shedding. In this review, we summarize evidence that cell surface receptor activity of PrPC is finely regulated by dimerization.
Collapse
Affiliation(s)
- Xavier Roucou
- Department of Biochemistry, Faculty of Medicine, Université de Sherbrooke Sherbrooke, QC, Canada
| |
Collapse
|
155
|
Shott RH, Appanah C, Grenier C, Tremblay G, Roucou X, Schang LM. Development of kinomic analyses to identify dysregulated signaling pathways in cells expressing cytoplasmic PrP. Virol J 2014; 11:175. [PMID: 25280966 PMCID: PMC4283144 DOI: 10.1186/1743-422x-11-175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/26/2014] [Indexed: 12/20/2022] Open
Abstract
Background Dysregulated protein kinase signaling is involved in the pathogenesis of many chronic diseases. However, the dysregulated signaling pathways critical to prion pathogenesis remain incompletely characterized. Global analyses of signaling pathways may be useful to better characterize these pathways. We therefore set out to develop such global assays. To this end, we used as a model cytoplasmic mutants of the cellular prion protein (PrPC), which are toxic to N2a neuroblastoma cells. We tested the global assays for their sensitivity to detect changes in signaling pathways in cells expressing cytoplasmic PrP mutants. Methods We developed a targeted proteomics (kinomics) approach using multiplex Western blots to identify signaling pathways dysregulated in chronic neurological pathologies. We tested the approach for its potential ability to detect signaling changes in N2a cells expressing cytoplasmic PrP mutants. Results Multiplex Western blots were designed to quantitate the expression levels of 137 protein kinases in a single membrane and using only 1.2 mg of sample. The response of the blots was sensitive and linear to changes of 6% in protein levels. Hierarchical and functional clustering of the relative expression levels identified an mTOR signaling pathway as potentially dysregulated in N2a cells expressing cytoplasmic PrP. The mTOR signaling pathway regulates global protein synthesis, which is inhibited in cells expressing cytoplasmic PrP. The levels of proteins involved in the Akt1/p70S6K branch of mTOR signaling changed in synchrony with time of cytoplasmic PrP expression. Three kinases in this pathway, Akt, p70S6K, and eIF4B were in their inactive states, as evaluated by phosphorylation of their regulatory sites. Conclusion The results presented are consistent with the previously reported inhibition of Akt/p70S6K/eIF4B signaling as mediating pathogenesis of cytoplasmic PrP. We conclude that the kinomic analyses are sensitive and specific to detect signaling pathways dysregulated in a simple in vitro model of PrP pathogenesis. Electronic supplementary material The online version of this article (doi:10.1186/1743-422X-11-175) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | - Luis M Schang
- Department of Biochemistry and Centre for Prions and Protein Folding Diseases (CPPFD), University of Alberta, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
156
|
Cichon AC, Brown DR. Nrf-2 regulation of prion protein expression is independent of oxidative stress. Mol Cell Neurosci 2014; 63:31-7. [PMID: 25242137 DOI: 10.1016/j.mcn.2014.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 08/18/2014] [Accepted: 09/12/2014] [Indexed: 12/13/2022] Open
Abstract
Cellular expression of host prion protein (PrP) is essential to infection with prion disease. Understanding the mechanisms that regulate prion protein expression at both the transcriptional and translational levels is therefore an important goal. The cellular prion protein has been associated with resistance to oxidative, and its expression is also increased by oxidative stress. The transcription factor Nrf-2 is associated with cellular responses to oxidative stress and is known to induce upregulation of antioxidant defense mechanisms. We have identified an Nrf-2 binding site in the prion protein promoter (Prnp) and shown that Nrf-2 downregulated PrP expression. However, this effect is independent of oxidative stress as oxidative stress can up-regulate PrP expression regardless of the level of Nrf-2 expression. Furthermore, Nrf-2 has no impact on PrP expression when cells are infected with scrapie. These findings highlight that Nrf-2 can regulate PrP expression, but that this regulation becomes uncoupled during cellular stress.
Collapse
Affiliation(s)
| | - David R Brown
- Department of Biology and Biochemistry, University of Bath, Bath, UK.
| |
Collapse
|
157
|
Song Z, Zhao D, Yang L. Metabolism of minor isoforms of prion proteins: Cytosolic prion protein and transmembrane prion protein. Neural Regen Res 2014; 8:2868-78. [PMID: 25206608 PMCID: PMC4146015 DOI: 10.3969/j.issn.1673-5374.2013.30.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 09/09/2013] [Indexed: 12/24/2022] Open
Abstract
Transmissible spongiform encephalopathy or prion disease is triggered by the conversion from cellular prion protein to pathogenic prion protein. Growing evidence has concentrated on prion protein configuration changes and their correlation with prion disease transmissibility and pathogenicity. In vivo and in vitro studies have shown that several cytosolic forms of prion protein with specific topological structure can destroy intracellular stability and contribute to prion protein pathogenicity. In this study, the latest molecular chaperone system associated with endoplasmic reticulum-associated protein degradation, the endoplasmic reticulum resident protein quality-control system and the ubiquitination proteasome system, is outlined. The molecular chaperone system directly correlates with the prion protein degradation pathway. Understanding the molecular mechanisms will help provide a fascinating avenue for further investigations on prion disease treatment and prion protein-induced neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhiqi Song
- State Key Laboratory for Agrobiotechnology, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Deming Zhao
- State Key Laboratory for Agrobiotechnology, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Lifeng Yang
- State Key Laboratory for Agrobiotechnology, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
158
|
Biochemical and immunological aspects of protein aggregation in neurodegenerative diseases. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2014. [DOI: 10.1007/s13738-014-0491-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
159
|
Abstract
Follicular dendritic cells (FDCs) are essential for high-affinity antibody production and for the development of B cell memory. Historically, FDCs have been characterized as 'accessory' cells that passively support germinal centre (GC) responses. However, recent observations suggest that FDCs actively shape humoral immunity. In this Review, we discuss recent findings concerning the antigen acquisition and retention functions of FDCs, and relevant implications for protective immunity. Furthermore, we describe the roles of FDCs within GCs in secondary lymphoid organs and discuss FDC development within this dynamic environment. Finally, we discuss how a better understanding of FDCs could facilitate the design of next-generation vaccines.
Collapse
|
160
|
Abstract
Neurodegenerative diseases are characterized by the aggregation of misfolded proteins in the brain. Among these disorders are the prion diseases, which are transmissible, and in which the misfolded proteins (“prions”) are also the infectious agent. Increasingly, it appears that misfolded proteins in Alzheimer and Parkinson diseases and the tauopathies also propagate in a “prion-like” manner. However, the association between prion formation, spread, and neurotoxicity is not clear. Recently, we showed that in prion disease, protein misfolding leads to neurodegeneration through dysregulation of generic proteostatic mechanisms, specifically, the unfolded protein response. Genetic and pharmacological manipulation of the unfolded protein response was neuroprotective despite continuing prion replication, hence dissociating this from neurotoxicity. The data have clear implications for treatment across the spectrum of these disorders, targeting pathogenic processes downstream of protein misfolding.
Collapse
Affiliation(s)
- Mark Halliday
- From the Medical Research Council (MRC) Toxicology Unit, Hodgkin Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Helois Radford
- From the Medical Research Council (MRC) Toxicology Unit, Hodgkin Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Giovanna R Mallucci
- From the Medical Research Council (MRC) Toxicology Unit, Hodgkin Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| |
Collapse
|
161
|
Abstract
The conformational diseases, linked to protein aggregation into amyloid conformations, range from non-infectious neurodegenerative disorders, such as Alzheimer disease (AD), to highly infectious ones, such as human transmissible spongiform encephalopathies (TSEs). They are commonly known as prion diseases. However, since all amyloids could be considered prions (from those involved in cell-to-cell transmission to those responsible for real neuronal invasion), it is necessary to find an underlying cause of the different capacity to infect that each of the proteins prone to form amyloids has. As proposed here, both the intrinsic cytotoxicity and the number of nuclei of aggregation per cell could be key factors in this transmission capacity of each amyloid.
Collapse
Affiliation(s)
- Raimon Sabate
- Conformational Diseases Group; Department of Physical Chemistry; Faculty of Pharmacy; University of Barcelona (UB); Barcelona, Spain; Institut of Nanoscience and Nanotechnology of the University of Barcelona (IN2UB); Barcelona, Spain
| |
Collapse
|
162
|
Abstract
Prion diseases are characterized by a conformational change in the normal host protein PrPC. While the majority of mature PrPC is tethered to the plasma membrane by a glycosylphosphatidylinositol anchor, topological variants of this protein can arise during its biosynthesis. Here we have generated Drosophila transgenic for cytosolic ovine PrP in order to investigate its toxic potential in flies in the absence or presence of exogenous ovine prions. While cytosolic ovine PrP expressed in Drosophila was predominantly detergent insoluble and showed resistance to low concentrations of proteinase K, it was not overtly detrimental to the flies. However, Drosophila transgenic for cytosolic PrP expression exposed to classical or atypical scrapie prion inocula showed a faster decrease in locomotor activity than similar flies exposed to scrapie-free material. The susceptibility to classical scrapie inocula could be assessed in Drosophila transgenic for panneuronal expression of cytosolic PrP, whereas susceptibility to atypical scrapie required ubiquitous PrP expression. Significantly, the toxic phenotype induced by ovine scrapie in cytosolic PrP transgenic Drosophila was transmissible to recipient PrP transgenic flies. These data show that while cytosolic PrP expression does not adversely affect Drosophila, this topological PrP variant can participate in the generation of transmissible scrapie-induced toxicity. These observations also show that PrP transgenic Drosophila are susceptible to classical and atypical scrapie prion strains and highlight the utility of this invertebrate host as a model of mammalian prion disease. Importance: During prion diseases, the host protein PrPC converts into an abnormal conformer, PrPSc, a process coupled to the generation of transmissible prions and neurotoxicity. While PrPC is principally a glycosylphosphatidylinositol-anchored membrane protein, the role of topological variants, such as cytosolic PrP, in prion-mediated toxicity and prion formation is undefined. Here we generated Drosophila transgenic for cytosolic PrP expression in order to investigate its toxic potential in the absence or presence of exogenous prions. Cytosolic ovine PrP expressed in Drosophila was not overtly detrimental to the flies. However, cytosolic PrP transgenic Drosophila exposed to ovine scrapie showed a toxic phenotype absent from similar flies exposed to scrapie-free material. Significantly, the scrapie-induced toxic phenotype in cytosolic transgenic Drosophila was transmissible to recipient PrP transgenic flies. These data show that cytosolic PrP can participate in the generation of transmissible prion-induced toxicity and highlight the utility of Drosophila as a model of mammalian prion disease.
Collapse
|
163
|
|
164
|
Cell-to-cell transmission of pathogenic proteins in neurodegenerative diseases. Nat Med 2014; 20:130-8. [PMID: 24504409 DOI: 10.1038/nm.3457] [Citation(s) in RCA: 494] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 12/19/2013] [Indexed: 12/12/2022]
Abstract
A common feature of many neurodegenerative diseases is the deposition of β-sheet-rich amyloid aggregates formed by proteins specific to these diseases. These protein aggregates are thought to cause neuronal dysfunction, directly or indirectly. Recent studies have strongly implicated cell-to-cell transmission of misfolded proteins as a common mechanism for the onset and progression of various neurodegenerative disorders. Emerging evidence also suggests the presence of conformationally diverse 'strains' of each type of disease protein, which may be another shared feature of amyloid aggregates, accounting for the tremendous heterogeneity within each type of neurodegenerative disease. Although there are many more questions to be answered, these studies have opened up new avenues for therapeutic interventions in neurodegenerative disorders.
Collapse
|
165
|
The inhibition of functional expression of calcium channels by prion protein demonstrates competition with α2δ for GPI-anchoring pathways. Biochem J 2014; 458:365-74. [PMID: 24329154 PMCID: PMC3924758 DOI: 10.1042/bj20131405] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
It has been shown recently that PrP (prion protein) and the calcium channel auxiliary α2δ subunits interact in neurons and expression systems [Senatore, Colleoni, Verderio, Restelli, Morini, Condliffe, Bertani, Mantovani, Canovi, Micotti, Forloni, Dolphin, Matteoli, Gobbi and Chiesa (2012) Neuron 74, 300-313]. In the present study we examined whether there was an effect of PrP on calcium currents. We have shown that when PrP is co-expressed with calcium channels formed from CaV2.1/β and α2δ-1 or α2δ-2, there is a consistent decrease in calcium current density. This reduction was absent when a PrP construct was used lacking its GPI (glycosylphosphatidylinositol) anchor. We have reported previously that α2δ subunits are able to form GPI-anchored proteins [Davies, Kadurin, Alvarez-Laviada, Douglas, Nieto-Rostro, Bauer, Pratt and Dolphin (2010) Proc. Natl. Acad. Sci. U.S.A. 107, 1654-1659] and show further evidence in the present paper. We have characterized recently a C-terminally truncated α2δ-1 construct, α2δ-1ΔC, and found that, despite loss of its membrane anchor, it still shows a partial ability to increase calcium currents [Kadurin, Alvarez-Laviada, Ng, Walker-Gray, D'Arco, Fadel, Pratt and Dolphin (2012) J. Biol. Chem. 1287, 33554-33566]. We now find that PrP does not inhibit CaV2.1/β currents formed with α2δ-1ΔC, rather than α2δ-1. It is possible that PrP and α2δ-1 compete for GPI-anchor intermediates or trafficking pathways, or that interaction between PrP and α2δ-1 requires association in cholesterol-rich membrane microdomains. Our additional finding that CaV2.1/β1b/α2δ-1 currents were inhibited by GPI-GFP, but not cytosolic GFP, indicates that competition for limited GPI-anchor intermediates or trafficking pathways may be involved in PrP suppression of α2δ subunit function.
Collapse
|
166
|
Johnson ML, Grazul-Bilska AT, Reynolds LP, Redmer DA. Prion (PrPC) expression in ovine uteroplacental tissues increases after estrogen treatment of ovariectomized ewes and during early pregnancy. Reproduction 2014; 148:1-10. [PMID: 24664411 DOI: 10.1530/rep-13-0548] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Scrapie in sheep is spread laterally by placental transmission of an infectious misfolded form (PrPSc) of a normal prion protein (PrPC) used as a template in PrPSc formation. We hypothesized that PrPC would be expressed in uterine and placental tissues and estradiol-17β (E2) would affect uterine PrPC expression. PrPC expression was evaluated in the uterus of long-term ovariectomized (OVX) ewes treated with an E2 implant for 2-24 h and in uteroplacental tissues from day 20 to day 30 of pregnancy. Expression of PrPC mRNA and PrPC protein increased in the uterus after E2 treatment of OVX ewes. In the maternal placenta, expression of PrPC mRNA and PrPC protein were unchanged, but in the fetal membranes (FM) PrPC mRNA and PrPC protein expression increased from day 20 to day 28. In the nonpregnant uterus, PrPC protein was immunolocalized at apical borders of the surface epithelium, in outer smooth muscle layers of large blood vessels, and in scattered stromal cells of the deep intercaruncular areas of the uterus. In the maternal placenta, PrPC protein was immunolocalized in the cytoplasm of flattened luminal epithelial cells apposed to the FM, whereas in the FM PrPC protein was in trophoblast cells and was also in several tissues of the developing embryo during early pregnancy. These data linking estrogen stimulation to increases in PrPC expression in uteroplacental tissues suggest that PrPC has a specific function during the estrous cycle and early pregnancy. Future studies should determine whether or not estrogen influences PrPC expression in other tissues, such as the nervous system and brain.
Collapse
Affiliation(s)
- Mary Lynn Johnson
- Department of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USADepartment of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USA
| | - Anna T Grazul-Bilska
- Department of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USADepartment of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USA
| | - Lawrence P Reynolds
- Department of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USADepartment of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USA
| | - Dale A Redmer
- Department of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USADepartment of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USA
| |
Collapse
|
167
|
Lara C, Reynolds NP, Berryman JT, Xu A, Zhang A, Mezzenga R. ILQINS Hexapeptide, Identified in Lysozyme Left-Handed Helical Ribbons and Nanotubes, Forms Right-Handed Helical Ribbons and Crystals. J Am Chem Soc 2014; 136:4732-9. [DOI: 10.1021/ja500445z] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Cecile Lara
- Food & Soft Materials, Department of Health Science & Technology, ETH Zurich, Schmelzbergstrasse 9, LFO, E23, 8092 Zürich, Switzerland
| | - Nicholas P. Reynolds
- Materials
Science and Engineering, CSIRO, Private Bag 10, Bayview Avenue, Clayton, Vic 3169, Australia
| | - Joshua T. Berryman
- Faculty
of Science Technology and Communication, University of Luxembourg, 162a Avenue de la Faïencerie, L-1511 Luxembourg
| | - Anqiu Xu
- Department
of Polymer Materials, Shanghai University, Nanchen Street 333, Shanghai 200444, China
| | - Afang Zhang
- Department
of Polymer Materials, Shanghai University, Nanchen Street 333, Shanghai 200444, China
| | - Raffaele Mezzenga
- Food & Soft Materials, Department of Health Science & Technology, ETH Zurich, Schmelzbergstrasse 9, LFO, E23, 8092 Zürich, Switzerland
| |
Collapse
|
168
|
Davidson L, Knight R. Neuropathogenesis of prion disease. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.13.74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT: Although much is known about prion diseases (characterized by a post-translational misfolding of the prion protein [PrP]) and their neuropathology and molecular pathology, the fundamental cause of illness, the basic neuropathogenesis, remains uncertain. There are three broad considerations discussed in this review: the possible loss of normal PrP function, the possible direct toxicity of the abnormally folded PrP and a harmful interaction between the normal and abnormal protein. In considering these possibilities, there are difficulties, including the facts that the relevant normal functions of the PrP are somewhat uncertain and that there are a number of possible toxic species of abnormal protein. In addition to the possible interactions of normal and abnormal PrP in prion disease, PrP may play a role in the neuropathogenesis of other diseases (such as Alzheimer’s disease).
Collapse
Affiliation(s)
- Louise Davidson
- National Creutzfeldt–Jakob Disease Research & Surveillance Unit, University of Edinburgh, Edinburgh, UK
| | - Richard Knight
- National Creutzfeldt–Jakob Disease Research & Surveillance Unit, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
169
|
Ning L, Guo J, Bai Q, Jin N, Liu H, Yao X. Structural diversity and initial oligomerization of PrP106-126 studied by replica-exchange and conventional molecular dynamics simulations. PLoS One 2014; 9:e87266. [PMID: 24586266 PMCID: PMC3929351 DOI: 10.1371/journal.pone.0087266] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 12/24/2013] [Indexed: 12/15/2022] Open
Abstract
Prion diseases are marked by cerebral accumulation of the abnormal isoform of the prion protein. A fragment of prion protein composed of residues 106–126 (PrP106–126) exhibits similar properties to full length prion and plays a key role in the conformational conversion from cellular prion to its pathogenic pattern. Soluble oligomers of PrP106–126 have been proposed to be responsible for neurotoxicity. However, the monomeric conformational space and initial oligomerization of PrP106–126 are still obscure, which are very important for understanding the conformational conversion of PrP106–126. In this study, replica exchange molecular dynamics simulations were performed to investigate monomeric and dimeric states of PrP106–126 in implicit solvent. The structural diversity of PrP106–126 was observed and this peptide did not acquire stable structure. The dimeric PrP106–126 also displayed structural diversity and hydrophobic interaction drove the dimerization. To further study initial oligomerization of PrP106–126, 1 µs conventional molecular dynamics simulations of trimer and tetramer formation were carried out in implicit solvent. We have observed the spontaneous formation of several basic oligomers and stable oligomers with high β-sheet contents were sampled in the simulations of trimer and tetramer formation. The β-hairpin formed in hydrophobic tail of PrP106–126 with residues 118–120 in turn may stabilize these oligomers and seed the formation oligomers. This study can provide insight into the detailed information about the structure of PrP106–126 and the dynamics of aggregation of monomeric PrP106–126 into oligomers in atomic level.
Collapse
Affiliation(s)
- Lulu Ning
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, China
| | - Jingjing Guo
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, China
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Qifeng Bai
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, China
| | | | - Huanxiang Liu
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, China
- School of Pharmacy, Lanzhou University, Lanzhou, China
- * E-mail: (HL); (XY)
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, China
- State Key Lab for Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
- * E-mail: (HL); (XY)
| |
Collapse
|
170
|
Mays CE, Kim C, Haldiman T, van der Merwe J, Lau A, Yang J, Grams J, Di Bari MA, Nonno R, Telling GC, Kong Q, Langeveld J, McKenzie D, Westaway D, Safar JG. Prion disease tempo determined by host-dependent substrate reduction. J Clin Invest 2014; 124:847-58. [PMID: 24430187 DOI: 10.1172/jci72241] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 11/07/2013] [Indexed: 01/01/2023] Open
Abstract
The symptoms of prion infection can take years or decades to manifest following the initial exposure. Molecular markers of prion disease include accumulation of the misfolded prion protein (PrPSc), which is derived from its cellular precursor (PrPC), as well as downregulation of the PrP-like Shadoo (Sho) glycoprotein. Given the overlapping cellular environments for PrPC and Sho, we inferred that PrPC levels might also be altered as part of a host response during prion infection. Using rodent models, we found that, in addition to changes in PrPC glycosylation and proteolytic processing, net reductions in PrPC occur in a wide range of prion diseases, including sheep scrapie, human Creutzfeldt-Jakob disease, and cervid chronic wasting disease. The reduction in PrPC results in decreased prion replication, as measured by the protein misfolding cyclic amplification technique for generating PrPSc in vitro. While PrPC downregulation is not discernible in animals with unusually short incubation periods and high PrPC expression, slowly evolving prion infections exhibit downregulation of the PrPC substrate required for new PrPSc synthesis and as a receptor for pathogenic signaling. Our data reveal PrPC downregulation as a previously unappreciated element of disease pathogenesis that defines the extensive, presymptomatic period for many prion strains.
Collapse
|
171
|
Lasmézas C, Zhou M. Newly defined toxic α-helical prion protein monomer: implications for other neurodegenerative diseases? Expert Rev Proteomics 2014; 9:233-5. [DOI: 10.1586/epr.12.26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
172
|
Qualtieri A, Urso E, Pera ML, Sprovieri T, Bossio S, Gambardella A, Quattrone A. Proteomic profiling of cerebrospinal fluid in Creutzfeldt–Jakob disease. Expert Rev Proteomics 2014; 7:907-17. [DOI: 10.1586/epr.10.80] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
173
|
Prion protein misfolding, strains, and neurotoxicity: an update from studies on Mammalian prions. Int J Cell Biol 2013; 2013:910314. [PMID: 24454379 PMCID: PMC3884631 DOI: 10.1155/2013/910314] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 11/10/2013] [Accepted: 11/11/2013] [Indexed: 11/17/2022] Open
Abstract
Prion diseases, also known as transmissible spongiform encephalopathies (TSEs), are a group of fatal neurodegenerative disorders affecting humans and other mammalian species. The central event in TSE pathogenesis is the conformational conversion of the cellular prion protein, PrPC, into the aggregate, β-sheet rich, amyloidogenic form, PrPSc. Increasing evidence indicates that distinct PrPSc conformers, forming distinct ordered aggregates, can encipher the phenotypic TSE variants related to prion strains. Prion strains are TSE isolates that, after inoculation into syngenic hosts, cause disease with distinct characteristics, such as incubation period, pattern of PrPSc distribution, and regional severity of histopathological changes in the brain. In analogy with other amyloid forming proteins, PrPSc toxicity is thought to derive from the existence of various intermediate structures prior to the amyloid fiber formation and/or their specific interaction with membranes. The latter appears particularly relevant for the pathogenesis of TSEs associated with GPI-anchored PrPSc, which involves major cellular membrane distortions in neurons. In this review, we update the current knowledge on the molecular mechanisms underlying three fundamental aspects of the basic biology of prions such as the putative mechanism of prion protein conversion to the pathogenic form PrPSc and its propagation, the molecular basis of prion strains, and the mechanism of induced neurotoxicity by PrPSc aggregates.
Collapse
|
174
|
Synaptic dysfunction in prion diseases: a trafficking problem? Int J Cell Biol 2013; 2013:543803. [PMID: 24369467 PMCID: PMC3863542 DOI: 10.1155/2013/543803] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/08/2013] [Indexed: 11/26/2022] Open
Abstract
Synaptic dysfunction is an important cause of neurological symptoms in prion diseases, a class of clinically heterogeneous neurodegenerative disorders caused by misfolding of the cellular prion protein (PrPC). Experimental data suggest that accumulation of misfolded PrPC in the endoplasmic reticulum (ER) may be crucial in synaptic failure, possibly because of the activation of the translational repression pathway of the unfolded protein response. Here, we report that this pathway is not operative in mouse models of genetic prion disease, consistent with our previous observation that ER stress is not involved. Building on our recent finding that ER retention of mutant PrPC impairs the secretory trafficking of calcium channels essential for synaptic function, we propose a model of pathogenicity in which intracellular retention of misfolded PrPC results in loss of function or gain of toxicity of PrPC-interacting proteins. This neurotoxic modality may also explain the phenotypic heterogeneity of prion diseases.
Collapse
|
175
|
p53 in neurodegenerative diseases and brain cancers. Pharmacol Ther 2013; 142:99-113. [PMID: 24287312 DOI: 10.1016/j.pharmthera.2013.11.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 11/07/2013] [Indexed: 12/21/2022]
Abstract
More than thirty years elapsed since a protein, not yet called p53 at the time, was detected to bind SV40 during viral infection. Thousands of papers later, p53 evolved as the main tumor suppressor involved in growth arrest and apoptosis. A lot has been done but the protein has not yet revealed all its secrets. Particularly important is the observation that in totally distinct pathologies where apoptosis is either exacerbated or impaired, p53 appears to play a central role. This is exemplified for Alzheimer's and Parkinson's diseases that represent the two main causes of age-related neurodegenerative affections, where cell death enhancement appears as one of the main etiological paradigms. Conversely, in cancers, about half of the cases are linked to mutations in p53 leading to the impairment of p53-dependent apoptosis. The involvement of p53 in these pathologies has driven a huge amount of studies aimed at designing chemical tools or biological approaches to rescue p53 defects or over-activity. Here, we describe the data linking p53 to neurodegenerative diseases and brain cancers, and we document the various strategies to interfere with p53 dysfunctions in these disorders.
Collapse
|
176
|
Onodera T, Sakudo A, Wu G, Saeki K. Bovine Spongiform Encephalopathy in Japan: History and Recent Studies on Oxidative Stress in Prion Diseases. Microbiol Immunol 2013; 50:565-78. [PMID: 16924141 DOI: 10.1111/j.1348-0421.2006.tb03831.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
With the respect to BSE and vCJD, compliance with the following three rules should strictly be observed: (i) Identification and destruction of all clinically affected cattle; (ii) destruction of all mammalian proteins used in feeding ruminant livestock; and (iii) destruction of all high-risk tissues for use in human consumption. Scrapie in sheep has been documented in the 18th century in the United Kingdom. Through studies of brain-to-brain transmission in the same species in 1935, Cuille et al. successfully isolated the culprit protein from the sheep brain. To transmit said protein from an animal to another, intracerebral inoculation was much more efficient than intraperitoneal or oral route in certain species; i.e. the hamster and mouse. Since discovery of the more efficacious infection route, studies and development of prion research have undergone 4 developmental phases. Phase I depicted discoveries of the pathological features of Creutzfeldt-Jakob disease (CJD) and scrapie with typical lesions of spongiform encephalopathy, while Phase II revealed individual-to-individual (or cross-species) transmissions of CJD, kuru and scrapie in animals. Phases I and II suggested the possible participation of a slow virus in the infection process. In Phase III, Prusiner et al. proposed the 'prion' theory in 1982, followed by the milestone development of the transgenic or gene-targeted mouse in prion research in Phase IV. By strain-typing of prions, CJD has been classified as type 2 or 4 by Parchi et al. and Wadsworth as type-2 or -4 and type-1 or -2, respectively. Wadsworth type 1 is detected in the cerebellum, while Wadsworth type 2 was detected in the prefrontal cortex of 10% of sporadic CJD patients. In 1999, Puoti et al. have reported the co-existence of two types of PrP(res) in a same patient. These reports indicated that PrP(res)-typing is a quantitative rather than a qualitative process, and the relationship between the molecular type and the prion strain is rather complex. In fact, previous findings of Truchot have correlated type-1 distribution with synaptic deposits, and type-2 with arrangement of diffuse deposits in neurons. Although the normal function of PrP(C) has not been fully understood, recent studies have shown that PrP(C) plays a role in copper metabolism, signal transduction, neuroprotection and cell maturation. Further search of PrP(C)-interacting molecules and detailed studies using Prnp(-/-) mice and various type of Prnp(-/-) cell lines under various conditions are the prerequisites in elucidating PrP functions. In the pathogenesis of prion diseases, present results support the hypothesis that 'loss-of-function' of PrP(C) decreases resistance to oxidative stress, and 'gain-of-function' of PrP(Sc) increases oxidative stress. The mechanisms of (i) the 'loss-of-function' of PrP(C) in enhanced susceptibility to oxidative stress and (ii) the 'gain-of-function' of PrP(Sc) in generation of oxidative stress remain to be elucidated, although their mechanisms of action, at least in part, involve the decrease and increase in SOD activity, respectively.
Collapse
Affiliation(s)
- Takashi Onodera
- Department of Molecular Immunology, School of Agricultural and Life Sciences, University of Tokyo
| | | | | | | |
Collapse
|
177
|
Convergence of synapses, endosomes, and prions in the biology of neurodegenerative diseases. Int J Cell Biol 2013; 2013:141083. [PMID: 24307901 PMCID: PMC3838826 DOI: 10.1155/2013/141083] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 09/23/2013] [Indexed: 11/17/2022] Open
Abstract
Age-related misfolding and aggregation of disease-linked proteins in selective brain regions is a characteristic of neurodegenerative diseases. Although neuropathological aggregates that characterize these various diseases are found at sites other than synapses, increasing evidence supports the idea that synapses are where the pathogenesis begins. Understanding these diseases is hampered by our lack of knowledge of what the normal functions of these proteins are and how they are affected by aging. Evidence has supported the idea that neurodegenerative disease-linked proteins have a common propensity for prion protein-like cell-to-cell propagation. However, it is not thought that the prion-like quality of these proteins/peptides that allows their cell-to-cell transmission implies a role for human-to-human spread in common age-related neurodegenerative diseases. It will be important to better understand the molecular and cellular mechanisms governing the role of these aggregating proteins in neural function, especially at synapses, how their propagation occurs and how pathogenesis is promoted by aging.
Collapse
|
178
|
Abstract
Individuals infected with prions succumb to brain damage, and prion infections continue to be inexorably lethal. However, many crucial steps in prion pathogenesis occur in lymphatic organs and precede invasion of the central nervous system. In the past two decades, a great deal has been learnt concerning the cellular and molecular mechanisms of prion lymphoinvasion. These properties are diagnostically useful and have, for example, facilitated preclinical diagnosis of variant Creutzfeldt-Jakob disease in the tonsils. Moreover, the early colonization of lymphoid organs can be exploited for post-exposure prophylaxis of prion infections. As stromal cells of lymphoid organs are crucial for peripheral prion infection, the dedifferentiation of these cells offers a powerful means of hindering prion spread in infected individuals. In this Review, we discuss the current knowledge of the immunobiology of prions with an emphasis on how basic discoveries might enable translational strategies.
Collapse
|
179
|
Tipper D, Martinez-Vilchez I, Markgren L, Kagalwala DZ. Mammalian Prion protein expression in yeast; a model for transmembrane insertion. Prion 2013; 7:477-87. [PMID: 24141197 DOI: 10.4161/pri.26850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The prion protein (PrP), a GPI-anchored glycoprotein, is inefficiently secreted by mammalian microsomes, 50% being found as transmembrane (TM) proteins with the central TM1 segment spanning the membrane. TM1 hydrophobicity is marginal for lateral membrane insertion, which is primarily driven by hydrophobic interaction between the ER translocon and substrates in transit. Most inserted TM1 has its N-terminus in the ER lumen (Ntm orientation), as expected for arrest of normal secretion. However, 20% is found in inverted Ctm orientation. These are minor species in vivo, presumably a consequence of efficient quality control. PrP mutations that increase TM1 hydrophobicity result in increased Ctm insertion, both in vitro and in mouse brain, and a strong correlation is found between CtmPrP insertion and neuropathology in transgenic mice; a copper-dependent pathogenicity mechanism is suggested. PrP fusions with a C-terminal epitope tag, when expressed in yeast cells at moderate levels, appear to interact efficiently with the translocon, providing a useful model for testing the effects of PrP mutations on TM insertion and orientation. However, secretion of PrP by the mammalian translocon requires the TRAP complex, absent in yeast, where essentially all PrP ends up as TM species, 85-90% Ntm and 10-15% Ctm. Although yeast is, therefore, an incomplete mimic of mammalian PrP trafficking, effects on Ctm insertion of mutations increasing TM1 hydrophobicity closely reflect those seen in vitro. Electrostatic substrate-translocon interactions are a major determinant of TM protein insertion orientation and the yeast model was used to investigate the role of the large negative charge difference across TM1, a likely cause of translocation delay that would favor TM insertion and Ctm orientation. An increase in ΔCh from -5 to -7 caused a marked increase in Ctm insertion, while a decrease to -3 or -1 allowed 35 and about 65% secretion, respectively. Utility of the yeast model and the role of this charge difference in driving PrP membrane insertion are confirmed.
Collapse
Affiliation(s)
- Donald Tipper
- Microbiology and Physiological Systems Department; University of Massachusetts Medical School; Worcester MA USA
| | | | - Lucas Markgren
- Mathematics Department; Doherty Memorial High School; Worcester MA USA
| | - Din Z Kagalwala
- Department of Anesthesiology; Hackensack University Medical Center; Hackensack, NJ USA
| |
Collapse
|
180
|
Fernández-Borges N, Eraña H, Elezgarai SR, Harrathi C, Gayosso M, Castilla J. Infectivity versus Seeding in Neurodegenerative Diseases Sharing a Prion-Like Mechanism. Int J Cell Biol 2013; 2013:583498. [PMID: 24187553 PMCID: PMC3800648 DOI: 10.1155/2013/583498] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/21/2013] [Indexed: 12/11/2022] Open
Abstract
Prions are considered the best example to prove that the biological information can be transferred protein to protein through a conformational change. The term "prion-like" is used to describe molecular mechanisms that share similarities with the mammalian prion protein self-perpetuating aggregation and spreading characteristics. Since prions are presumably composed only of protein and are infectious, the more similar the mechanisms that occur in the different neurodegenerative diseases, the more these processes will resemble an infection. In vitro and in vivo experiments carried out during the last decade in different neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's diseases (PD), and amyotrophic lateral sclerosis (ALS) have shown a convergence toward a unique mechanism of misfolded protein propagation. In spite of the term "infection" that could be used to explain the mechanism governing the diversity of the pathological processes, other concepts as "seeding" or "de novo induction" are being used to describe the in vivo propagation and transmissibility of misfolded proteins. The current studies are demanding an extended definition of "disease-causing agents" to include those already accepted as well as other misfolded proteins. In this new scenario, "seeding" would be a type of mechanism by which an infectious agent can be transmitted but should not be used to define a whole "infection" process.
Collapse
Affiliation(s)
| | - Hasier Eraña
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, 48160 Bizkaia, Spain
| | | | - Chafik Harrathi
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, 48160 Bizkaia, Spain
| | - Mayela Gayosso
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, 48160 Bizkaia, Spain
| | - Joaquín Castilla
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, 48160 Bizkaia, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, 48011 Bizkaia, Spain
| |
Collapse
|
181
|
Sonati T, Reimann RR, Falsig J, Baral PK, O'Connor T, Hornemann S, Yaganoglu S, Li B, Herrmann US, Wieland B, Swayampakula M, Rahman MH, Das D, Kav N, Riek R, Liberski PP, James MNG, Aguzzi A. The toxicity of antiprion antibodies is mediated by the flexible tail of the prion protein. Nature 2013; 501:102-6. [PMID: 23903654 DOI: 10.1038/nature12402] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 06/21/2013] [Indexed: 12/17/2022]
Abstract
Prion infections cause lethal neurodegeneration. This process requires the cellular prion protein (PrP(C); ref. 1), which contains a globular domain hinged to a long amino-proximal flexible tail. Here we describe rapid neurotoxicity in mice and cerebellar organotypic cultured slices exposed to ligands targeting the α1 and α3 helices of the PrP(C) globular domain. Ligands included seven distinct monoclonal antibodies, monovalent Fab1 fragments and recombinant single-chain variable fragment miniantibodies. Similar to prion infections, the toxicity of globular domain ligands required neuronal PrP(C), was exacerbated by PrP(C) overexpression, was associated with calpain activation and was antagonized by calpain inhibitors. Neurodegeneration was accompanied by a burst of reactive oxygen species, and was suppressed by antioxidants. Furthermore, genetic ablation of the superoxide-producing enzyme NOX2 (also known as CYBB) protected mice from globular domain ligand toxicity. We also found that neurotoxicity was prevented by deletions of the octapeptide repeats within the flexible tail. These deletions did not appreciably compromise globular domain antibody binding, suggesting that the flexible tail is required to transmit toxic signals that originate from the globular domain and trigger oxidative stress and calpain activation. Supporting this view, various octapeptide ligands were not only innocuous to both cerebellar organotypic cultured slices and mice, but also prevented the toxicity of globular domain ligands while not interfering with their binding. We conclude that PrP(C) consists of two functionally distinct modules, with the globular domain and the flexible tail exerting regulatory and executive functions, respectively. Octapeptide ligands also prolonged the life of mice expressing the toxic PrP(C) mutant, PrP(Δ94-134), indicating that the flexible tail mediates toxicity in two distinct PrP(C)-related conditions. Flexible tail-mediated toxicity may conceivably play a role in further prion pathologies, such as familial Creutzfeldt-Jakob disease in humans bearing supernumerary octapeptides.
Collapse
Affiliation(s)
- Tiziana Sonati
- Institute of Neuropathology, University Hospital Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Boese AS, Majer A, Saba R, Booth SA. Small RNA drugs for prion disease: a new frontier. Expert Opin Drug Discov 2013; 8:1265-84. [DOI: 10.1517/17460441.2013.818976] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
183
|
The Functional Role of Prion Protein (PrPC) on Autophagy. Pathogens 2013; 2:436-45. [PMID: 25437200 PMCID: PMC4235692 DOI: 10.3390/pathogens2030436] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/11/2013] [Accepted: 06/18/2013] [Indexed: 12/19/2022] Open
Abstract
Cellular prion protein (PrPC) plays an important role in the cellular defense against oxidative stress. However, the exact protective mechanism of PrPC is unclear. Autophagy is essential for survival, differentiation, development, and homeostasis in several organisms. Although the role that autophagy plays in neurodegenerative disease has yet to be established, it is clear that autophagy-induced cell death is observed in neurodegenerative disorders that exhibit protein aggregations. Moreover, autophagy can promote cell survival and cell death under various conditions. In this review, we describe the involvement of autophagy in prion disease and the effects of PrPC.
Collapse
|
184
|
Using protein misfolding cyclic amplification generates a highly neurotoxic PrP dimer causing neurodegeneration. J Mol Neurosci 2013; 51:655-62. [PMID: 23771785 DOI: 10.1007/s12031-013-0039-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 05/27/2013] [Indexed: 10/26/2022]
Abstract
Under the "protein-only" hypothesis, prion-based diseases are proposed to result from an infectious agent that is an abnormal isoform of the prion protein in the scrapie form, PrP(Sc). However, since PrP(Sc) is highly insoluble and easily aggregates in vivo, this view appears to be overly simplistic, implying that the presence of PrP(Sc) may indirectly cause neurodegeneration through its intermediate soluble form. We generated a neurotoxic PrP dimer with partial pathogenic characteristics of PrP(Sc) by protein misfolding cyclic amplification in the presence of 1-palmitoyl-2-oleoylphosphatidylglycerol consisting of recombinant hamster PrP (23-231). After intracerebral injection of the PrP dimer, wild-type hamsters developed signs of neurodegeneration. Clinical symptoms, necropsy findings, and histopathological changes were very similar to those of transmissible spongiform encephalopathies. Additional investigation showed that the toxicity is primarily related to cellular apoptosis. All results suggested that we generated a new neurotoxic form of PrP, PrP dimer, which can cause neurodegeneration. Thus, our study introduces a useful model for investigating PrP-linked neurodegenerative mechanisms.
Collapse
|
185
|
Hu PP, Huang CZ. Prion protein: structural features and related toxicity. Acta Biochim Biophys Sin (Shanghai) 2013; 45:435-41. [PMID: 23615535 DOI: 10.1093/abbs/gmt035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Transmissible spongiform encephalopathies, or prion diseases, is a group of infectious neurodegenerative disorders. The conformational conversion from cellular form (PrP(C)) to disease-causing isoform (PrP(Sc)) is considered to be the most important and remarkable event in these diseases, while accumulation of PrP(Sc) is thought to be the main reason for cell death, inflammation and spongiform degeneration observed in infected individuals. Although these rare but unique neurodegenerative disorders have attracted much attention, there are still many questions that remain to be answered. Knowledge of the scrapie agent structures and the toxic species may have significance for understanding the causes of the diseases, and could be helpful for rational design of novel therapeutic and diagnostic methods. In this review, we summarized the available experimental evidence concerning the relationship among the structural features, aggregation status of misfolded PrP and related neurotoxicity in the course of prion diseases development. In particular, most data supports the idea that the smaller oligomeric PrP(Sc) aggregates, rather than the mature amyloid fibers, exhibit the highest toxicity to the host.
Collapse
Affiliation(s)
- Ping Ping Hu
- Ministry of Education Key Laboratory on Luminescence and Real-Time Analysis, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | | |
Collapse
|
186
|
Kretzschmar H, Tatzelt J. Prion disease: a tale of folds and strains. Brain Pathol 2013; 23:321-32. [PMID: 23587138 PMCID: PMC8029118 DOI: 10.1111/bpa.12045] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 02/04/2013] [Indexed: 12/31/2022] Open
Abstract
Research on prions, the infectious agents of devastating neurological diseases in humans and animals, has been in the forefront of developing the concept of protein aggregation diseases. Prion diseases are distinguished from other neurodegenerative diseases by three peculiarities. First, prion diseases, in addition to being sporadic or genetic like all other neurodegenerative diseases, are infectious diseases. Animal models were developed early on (a long time before the advent of transgenic technology), and this has made possible the discovery of the prion protein as the infectious agent. Second, human prion diseases have true equivalents in animals, such as scrapie, which has been the subject of experimental research for many years. Variant Creutzfeldt-Jakob disease (vCJD) is a zoonosis caused by bovine spongiform encephalopathy (BSE) prions. Third, they show a wide variety of phenotypes in humans and animals, much wider than the variants of any other sporadic or genetic neurodegenerative disease. It has now become firmly established that particular PrP(Sc) isoforms are closely related to specific human prion strains. The variety of human prion diseases, still an enigma in its own right, is a focus of this article. Recently, a series of experiments has shown that the concept of aberrant protein folding and templating, first developed for prions, may apply to a variety of neurodegenerative diseases. In the wake of these discoveries, the term prion has come to be used for Aβ, α-synuclein, tau and possibly others. The self-propagation of alternative conformations seems to be the common denominator of these "prions," which in future, in order to avoid confusion, may have to be specified either as "neurodegenerative prions" or "infectious prions."
Collapse
Affiliation(s)
| | - Jörg Tatzelt
- NeurobiochemistryAdolf‐Butenandt‐InstituteLudwig‐Maximilians‐University MunichMunichGermany
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| |
Collapse
|
187
|
Zhou J, Liu B. Alzheimer's disease and prion protein. Intractable Rare Dis Res 2013; 2:35-44. [PMID: 25343100 PMCID: PMC4204584 DOI: 10.5582/irdr.2013.v2.2.35] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 05/20/2013] [Accepted: 05/22/2013] [Indexed: 11/05/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease with progressive loss of memory and cognitive function, pathologically hallmarked by aggregates of the amyloid-beta (Aβ) peptide and hyperphosphorylated tau in the brain. Aggregation of Aβ under the form of amyloid fibrils has long been considered central to the pathogenesis of AD. However, recent evidence has indicated that soluble Aβ oligomers, rather than insoluble fibrils, are the main neurotoxic species in AD. The cellular prion protein (PrP(C)) has newly been identified as a cell surface receptor for Aβ oligomers. PrP(C) is a cell surface glycoprotein that plays a key role in the propagation of prions, proteinaceous infectious agents that replicate by imposing their abnormal conformation to PrP(C) molecules. In AD, PrP(C) acts to transduce the neurotoxic signals arising from Aβ oligomers, leading to synaptic failure and cognitive impairment. Interestingly, accumulating evidence has also shown that aggregated Aβ or tau possesses prion-like activity, a property that would allow them to spread throughout the brain. In this article, we review recent findings regarding the function of PrP(C) and its role in AD, and discuss potential therapeutic implications of PrP(C)-based approaches in the treatment of AD.
Collapse
Affiliation(s)
- Jiayi Zhou
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
- Address correspondence to: Dr. Jiayi Zhou, Department of Biochemistry, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA. E-mail:
| | - Bingqian Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
188
|
Abstract
Growing evidence suggests that a physiological activity of the cellular prion protein (PrP(C)) plays a crucial role in several neurodegenerative disorders, including prion and Alzheimer's diseases. However, how the functional activity of PrP(C) is subverted to deliver neurotoxic signals remains uncertain. Transgenic (Tg) mice expressing PrP with a deletion of residues 105-125 in the central region (referred to as ΔCR PrP) provide important insights into this problem. Tg(ΔCR) mice exhibit neonatal lethality and massive degeneration of cerebellar granule neurons, a phenotype that is dose dependently suppressed by the presence of wild-type PrP. When expressed in cultured cells, ΔCR PrP induces large, ionic currents that can be detected by patch-clamping techniques. Here, we tested the hypothesis that abnormal ion channel activity underlies the neuronal death seen in Tg(ΔCR) mice. We find that ΔCR PrP induces abnormal ionic currents in neurons in culture and in cerebellar slices and that this activity sensitizes the neurons to glutamate-induced, calcium-mediated death. In combination with ultrastructural and biochemical analyses, these results demonstrate a role for glutamate-induced excitotoxicity in PrP-mediated neurodegeneration. A similar mechanism may operate in other neurodegenerative disorders attributable to toxic, β-rich oligomers that bind to PrP(C).
Collapse
|
189
|
Uppington KM, Brown DR. Modelling neurodegeneration in prion disease - applications for drug development. Expert Opin Drug Discov 2013; 2:777-88. [PMID: 23488996 DOI: 10.1517/17460441.2.6.777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Prion diseases are a group of neurodegenerative diseases that affect mammals, including humans and ruminants such as sheep. They are believed to be caused by the conversion of the prion protein (PrP), a host expressed protein, into a toxic form (PrP(sc)). PrP(sc) accumulates in the brain, resulting in neuronal loss and the typical spongiform appearance of the brain. So far, there are no effective therapies available for prion diseases. This review discusses possible therapies for prion diseases and the models available for advancing research into the disease.
Collapse
Affiliation(s)
- Kay M Uppington
- University of Bath, Department of Biology and Biochemistry, Bath, Claverton Down, BA2 7AY, UK +44 1255 383133 ; +44 1225 386779 ;
| | | |
Collapse
|
190
|
Altmeppen HC, Prox J, Puig B, Dohler F, Falker C, Krasemann S, Glatzel M. Roles of endoproteolytic α-cleavage and shedding of the prion protein in neurodegeneration. FEBS J 2013; 280:4338-47. [DOI: 10.1111/febs.12196] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/25/2013] [Accepted: 02/14/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Hermann C. Altmeppen
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| | - Johannes Prox
- Institute of Biochemistry; Christian Albrechts University; Kiel Germany
| | - Berta Puig
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| | - Frank Dohler
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| | - Clemens Falker
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| | - Susanne Krasemann
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| | - Markus Glatzel
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| |
Collapse
|
191
|
Huang X, Long Y, Zhang H, Wang Q, Zhu R, Zheng H. Gold nanoparticles as a probe for prion determination via resonance light scattering method. ANAL SCI 2013; 28:475-9. [PMID: 22687927 DOI: 10.2116/analsci.28.475] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Dihydrolipoic acid (DHLA) modified gold nanoparticles (AuNPs) were used as a highly selective probe for the detection of prion proteins. We discovered that AuNPs undergo aggregation selectively in the presence of recombinant prion protein (rPrP), and such selective aggregation enhanced the resonance light scattering (RLS) intensity from AuNPs tremendously. Based on this phenomenon, we established a new assay for rPrP detection. This new assay is label-free, highly selective, and sensitive. The linear range for rPrP detection is from 2.0 × 10(-10) to 2.0 × 10(-8) mol L(-1) with excellent discrimination against other interfering compounds, and the detection limit is 7 × 10(-11) mol L(-1). This assay has been successfully applied for rPrP detection in E. coli lysate, bovine serum samples and human plasma samples. Compared with other methods, the detection approach described here can achieve high selectivity and sensitivity without any complicated labeling or expensive instruments.
Collapse
Affiliation(s)
- XiaoXiao Huang
- Key Laboratory on Luminescence and Real-Time Analysis, Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Beibei, Chongqing, PR China
| | | | | | | | | | | |
Collapse
|
192
|
Head MW. Human prion diseases: Molecular, cellular and population biology. Neuropathology 2013; 33:221-36. [DOI: 10.1111/neup.12016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/17/2012] [Indexed: 12/17/2022]
Affiliation(s)
- Mark W. Head
- National CJD Research & Surveillance Unit; Centre for Clinical Brain Sciences; School of Clinical Sciences; The University of Edinburgh; Edinburgh; UK
| |
Collapse
|
193
|
|
194
|
Resenberger UK, Müller V, Munter LM, Baier M, Multhaup G, Wilson MR, Winklhofer KF, Tatzelt J. The heat shock response is modulated by and interferes with toxic effects of scrapie prion protein and amyloid β. J Biol Chem 2012; 287:43765-76. [PMID: 23115236 PMCID: PMC3527961 DOI: 10.1074/jbc.m112.389007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 10/27/2012] [Indexed: 12/19/2022] Open
Abstract
The heat shock response (HSR) is an evolutionarily conserved pathway designed to maintain proteostasis and to ameliorate toxic effects of aberrant protein folding. We have studied the modulation of the HSR by the scrapie prion protein (PrP(Sc)) and amyloid β peptide (Aβ) and investigated whether an activated HSR or the ectopic expression of individual chaperones can interfere with PrP(Sc)- or Aβ-induced toxicity. First, we observed different effects on the HSR under acute or chronic exposure of cells to PrP(Sc) or Aβ. In chronically exposed cells the threshold to mount a stress response was significantly increased, evidenced by a decreased expression of Hsp72 after stress, whereas an acute exposure lowered the threshold for stress-induced expression of Hsp72. Next, we employed models of PrP(Sc)- and Aβ-induced toxicity to demonstrate that the induction of the HSR ameliorates the toxic effects of both PrP(Sc) and Aβ. Similarly, the ectopic expression of cytosolic Hsp72 or the extracellular chaperone clusterin protected against PrP(Sc)- or Aβ-induced toxicity. However, toxic signaling induced by a pathogenic PrP mutant located at the plasma membrane was prevented by an activated HSR or Hsp72 but not by clusterin, indicating a distinct mode of action of this extracellular chaperone. Our study supports the notion that different pathological protein conformers mediate toxic effects via similar cellular pathways and emphasizes the possibility to exploit the heat shock response therapeutically.
Collapse
Affiliation(s)
- Ulrike K. Resenberger
- From the Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University Munich, D-80336 Munich, Germany
| | - Veronika Müller
- From the Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University Munich, D-80336 Munich, Germany
| | - Lisa M. Munter
- Institut für Chemie und Biochemie, Freie Universität, 14195 Berlin, Germany
- the Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3A0G4, Canada
| | | | - Gerd Multhaup
- Institut für Chemie und Biochemie, Freie Universität, 14195 Berlin, Germany
- the Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3A0G4, Canada
| | - Mark R. Wilson
- the School of Biological Sciences, University of Wollongong, Wollongong, New South Wales 2522, Australia, and
| | - Konstanze F. Winklhofer
- From the Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University Munich, D-80336 Munich, Germany
- the German Center for Neurodegenerative Diseases (DZNE), 80336 Munich, Germany
| | - Jörg Tatzelt
- From the Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University Munich, D-80336 Munich, Germany
- the German Center for Neurodegenerative Diseases (DZNE), 80336 Munich, Germany
| |
Collapse
|
195
|
PrP(C) homodimerization stimulates the production of PrPC cleaved fragments PrPN1 and PrPC1. J Neurosci 2012; 32:13255-63. [PMID: 22993441 DOI: 10.1523/jneurosci.2236-12.2012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
An endoproteolytic cleavage termed α-cleavage between residues 111/112 is a characteristic feature of the cellular prion protein (PrP(C)). This cleavage generates a soluble N-terminal fragment (PrPN1) and a glycosylphosphatidylinositol-anchored C-terminal fragment (PrPC1). Independent studies demonstrate that modulating PrP(C) α-cleavage represents a potential therapeutic strategy in prion diseases. The regulation of PrP(C) α-cleavage is unclear. The only known domain that is essential for the α-cleavage to occur is a hydrophobic domain (HD). Importantly, the HD is also essential for the formation of PrP(C) homodimers. To explore the role of PrP(C) homodimerization on the α-cleavage, we used a well described inducible dimerization strategy whereby a chimeric PrP(C) composed of a modified FK506-binding protein (Fv) fused with PrP(C) and termed Fv-PrP is incubated in the presence of a dimerizer AP20187 ligand. We show that homodimerization leads to a considerable increase of PrP(C) α-cleavage in cultured cells and release of PrPN1 and PrPC1. Interestingly, enforced homodimerization increased PrP(C) levels at the plasma membrane, and preventing PrP(C) trafficking to the cell surface inhibited dimerization-induced α-cleavage. These observations were confirmed in primary hippocampal neurons from transgenic mice expressing Fv-PrP. The proteases responsible for the α-cleavage are still elusive, and in contrast to initial studies we confirm more recent investigations that neither ADAM10 nor ADAM17 are involved. Importantly, PrPN1 produced after PrP(C) homodimerization protects against toxic amyloid-β (Aβ) oligomers. Thus, our results show that PrP(C) homodimerization is an important regulator of PrP(C) α-cleavage and may represent a potential therapeutic avenue against Aβ toxicity in Alzheimer's disease.
Collapse
|
196
|
Abstract
Like patients with prion disease, Alzheimer patients suffer from a fatal, progressive form of dementia. There is growing evidence that amyloid-β (Aβ) aggregates may be transmissible similar to prions, at least under extreme experimental conditions. However, unlike mice infected with prion protein (PrP) prions, those inoculated with Aβ do not die. The transmission of Aβ and PrP thus differs conspicuously in the neurological effects they induce in their hosts, the difference being no less than a matter of life and death. Far from being a mere academic nuance, this distinction between Aβ and PrP begs the crucial questions of what, exactly, controls prion toxicity and how prion toxicity relates to prion infectivity.
Collapse
Affiliation(s)
- Karen H Ashe
- Department of Neurology, N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA.
| | | |
Collapse
|
197
|
Abstract
The human cellular prion protein (PrP(C)) is a glycosylphosphatidylinositol (GPI) anchored membrane glycoprotein with two N-glycosylation sites at residues 181 and 197. This protein migrates in several bands by Western blot analysis (WB). Interestingly, PNGase F treatment of human brain homogenates prior to the WB, which is known to remove the N-glycosylations, unexpectedly gives rise to two dominant bands, which are now known as C-terminal (C1) and N-terminal (N1) fragments. This resembles the β-amyloid precursor protein (APP) in Alzheimer disease (AD), which can be physiologically processed by α-, β-, and γ-secretases. The processing of APP has been extensively studied, while the identity of the cellular proteases involved in the proteolysis of PrP(C) and their possible role in prion biology has remained limited and controversial. Nevertheless, there is a strong correlation between the neurotoxicity caused by prion proteins and the blockade of their normal proteolysis. For example, expression of non-cleavable PrP(C) mutants in transgenic mice generates neurotoxicity, even in the absence of infectious prions, suggesting that PrP(C) proteolysis is physiologically and pathologically important. As many mouse models of prion diseases have recently been developed and the knowledge about the proteases responsible for the PrP(C) proteolysis is accumulating, we examine the historical experimental evidence and highlight recent studies that shed new light on this issue.
Collapse
|
198
|
Abstract
Transmissible spongiform encephalopathies (TSEs) are fatal neurodegenerative diseases that occur in a wide variety of mammals. In humans, TSE diseases include kuru, sporadic and iatrogenic Creutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker syndrome (GSS), and fatal familial insomnia (FFI). So far, TSE diseases occur only rarely in humans; however, scrapie is a widespread problem in sheep, and the recent epidemic of bovine spongiform encephalopathy (BSE or mad cow disease) has seriously affected the British cattle industry. Of special concern is the recent appearance of a new variant of CJD in humans that is suspected of being caused by infections from BSE-infected cattle products. In all these diseases, an abnormal form of a host protein, prion protein (PrP), is essential for the pathogenic process. The relationship of this protein to the transmissible agent is currently the subject of great interest and controversy and is the subject of this review.
Collapse
|
199
|
Solomon IH, Biasini E, Harris DA. Ion channels induced by the prion protein: mediators of neurotoxicity. Prion 2012; 6:40-5. [PMID: 22453177 DOI: 10.4161/pri.6.1.18627] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Prion diseases comprise a group of rapidly progressive and invariably fatal neurodegenerative disorders for which there are no effective treatments. While conversion of the cellular prion protein (PrP(C)) to a β-sheet rich isoform (PrP(Sc) ) is known to be a critical event in propagation of infectious prions, the identity of the neurotoxic form of PrP and its mechanism of action remain unclear. Insights into this mechanism have been provided by studying PrP molecules harboring deletions and point mutations in the conserved central region, encompassing residues 105-125. When expressed in transgenic mice, PrP deleted for these residues (Δ105-125) causes a spontaneous neurodegenerative illness that is reversed by co-expression of wild-type PrP. In cultured cells, Δ105-125 PrP confers hypersensitivity to certain cationic antibiotics and induces spontaneous ion channel activity that can be recorded by electrophysiological techniques. We have utilized these drug-hypersensitization and current-inducing activities to identify which PrP domains and subcellular locations are required for toxicity. We present an ion channel model for the toxicity of Δ105-125 PrP and related mutants and speculate how a similar mechanism could mediate PrP(Sc)-associated toxicity. Therapeutic regimens designed to inhibit prion-induced toxicity, as well as formation of PrP(Sc) , may prove to be the most clinically beneficial.
Collapse
Affiliation(s)
- Isaac H Solomon
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | | | | |
Collapse
|
200
|
Krasemann S, Neumann M, Luepke JP, Grashorn J, Wurr S, Stocking C, Glatzel M. Persistent retroviral infection with MoMuLV influences neuropathological signature and phenotype of prion disease. Acta Neuropathol 2012; 124:111-26. [PMID: 22271154 DOI: 10.1007/s00401-012-0944-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 01/08/2012] [Accepted: 01/09/2012] [Indexed: 12/26/2022]
Abstract
A fundamental step in pathophysiology of prion diseases is the conversion of the host encoded prion protein (PrP(C)) into a misfolded isoform (PrP(Sc)) that accumulates mainly in neuronal but also non-neuronal tissues. Prion diseases are transmissible within and between species. In a subset of prion diseases, peripheral prion uptake and subsequent transport to the central nervous system are key to disease initiation. The involvement of retroviruses in this process has been postulated based on the findings that retroviral infections enhance the spread of prion infectivity and PrP(Sc) from cell to cell in vitro. To study whether retroviral infection influences the phenotype of prion disease or the spread of prion infectivity and PrP(Sc) in vivo, we developed a murine model with persistent Moloney murine leukemia retrovirus (MoMuLV) infection with and without additional prion infection. We investigated the pathophysiology of prion disease in MoMuLV and prion-infected mice, monitoring temporal kinetics of PrP(Sc) spread and prion infectivity, as well as clinical presentation. Unexpectedly, infection of MoMuLV challenged mice with prions did not change incubation time to clinical prion disease. However, clinical presentation of prion disease was altered in mice infected with both pathogens. This was paralleled by remarkably enhanced astrogliosis and pathognomonic astrocyte morphology in the brain of these mice. Therefore, we conclude that persistent viral infection might act as a disease modifier in prion disease.
Collapse
Affiliation(s)
- Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|