1
|
Juliani do Amaral M, Soares de Oliveira L, Cordeiro Y. Zinc ions trigger the prion protein liquid-liquid phase separation. Biochem Biophys Res Commun 2025; 753:151489. [PMID: 39983547 DOI: 10.1016/j.bbrc.2025.151489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Abstract
Prion diseases are characterized by the misfolding and conversion of the monomeric prion protein (PrP) to a multimeric aggregated pathogenic form, known as PrPSc. We and others have recently shown that biomolecular condensates formed via liquid-liquid phase separation of PrP can undergo maturation to solid-like species that resemble pathological aggregates, and this process is modulated by DNA, RNA, and oxidative conditions. Conversely, the most well-studied ligand of PrP, copper ions, induce liquid-like condensates of PrP that accumulate Cu2+in vitro, and live PrPC-expressing cells show condensation at the cell surface as triggered by physiologically relevant conditions of Cu2+ and protein concentrations. Since PrP can also bind to Zn2+ through its intrinsically disordered N-terminal domain, though with different affinities and binding modes than Cu2+, we hypothesized that Zn2+ could modulate PrP phase separation differently from copper ions. Using an appropriate buffer with negligible metal ion binding, as well as relevant pH, ionic strength, molecular crowding, and Zn2+ concentrations, we show that recombinant PrP undergoes phase separation with Zn2+. Furthermore, we show that metal ion-induced condensation of PrP is dependent on the N-terminal domain (residues 23-90). In vitro Fluorescence Recovery After Photobleaching (FRAP) experiments and thioflavin T aggregation kinetics support key differences in the molecular properties of PrP:Zn2+versus PrP:Cu2+ phase separated states. FRAP analysis indicated that both Cu2+ and Zn2+ promote liquid-like PrP condensates; however, PrP:Zn2+condensates exhibit a faster recovery. Cu2+ pronouncedly inhibits seed-induced PrP misfolding, whereas Zn2+ provides a milder delay in PrP aggregation. Our findings provide insights on Zn2+-induced phase separation of PrP, supporting a variety of previously proposed functions of PrP in metal sequestering and uptake, processes that could be effectively regulated through biomolecular condensation.
Collapse
Affiliation(s)
| | | | - Yraima Cordeiro
- Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Pehar M, Hewitt M, Wagner A, Sandhu JK, Khalili A, Wang X, Cho JY, Sim VL, Kulka M. Histamine stimulates human microglia to alter cellular prion protein expression via the HRH2 histamine receptor. Sci Rep 2024; 14:25519. [PMID: 39462031 PMCID: PMC11513956 DOI: 10.1038/s41598-024-75982-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Although the cellular prion protein (PrPC) has been evolutionarily conserved, the role of this protein remains elusive. Recent evidence indicates that PrPC may be involved in neuroinflammation and the immune response in the brain, and its expression may be modified via various mechanisms. Histamine is a proinflammatory mediator and neurotransmitter that stimulates numerous cells via interactions with histamine receptors 1-4 (HRH1-4). Since microglia are the innate immune cells of the central nervous system, we hypothesized that histamine-induced stimulation regulates the expression of PrPC in human-derived microglia. The human microglial clone 3 (HMC3) cell line was treated with histamine, and intracellular calcium levels were measured via a calcium flux assay. Cytokine production was monitored by enzyme-linked immunosorbent assay (ELISA). Western blotting and quantitative reverse transcription-polymerase chain reaction were used to determine protein and gene expression of HRH1-4. Flow cytometry and western blotting were used to measure PrPC expression levels. Fluorescence microscopy was used to examine Iba-1 and PrPC localization. HMC3 cells stimulated by histamine exhibited increased intracellular calcium levels and increased release of IL-6 and IL-8, while also modifying PrPC localization. HMC3 stimulated with histamine for 6 and 24 hours exhibited increased surface PrPC expression. Specifically, we found that stimulation of the HRH2 receptor was responsible for changes in surface PrPC. Histamine-induced increases in surface PrPC were attenuated following inhibition of the HRH2 receptor via the HRH2 antagonist ranitidine. These changes were unique to HRH2 activation, as stimulation of HRH1, HRH3, or HRH4 did not alter surface PrPC. Prolonged stimulation of HMC3 decreased PrPC expression following 48 and 72 hours of histamine stimulation. HMC3 cells can be stimulated by histamine to undergo intracellular calcium influx. Surface expression levels of PrPC on HMC3 cells are altered by histamine exposure, primarily mediated by HRH2. While histamine exposure also increases release of IL-6 and IL-8 in these cells, this cytokine release is not fully dependent on PrPC levels, as IL-6 release is only partially reduced and IL-8 release is unchanged under the conditions of HRH2 blockade that prevent PrPC changes. Overall, this suggests that PrPC may play a role in modulating microglial responses.
Collapse
Affiliation(s)
- Marcus Pehar
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - Melissa Hewitt
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Ashley Wagner
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
| | - Jagdeep K Sandhu
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Aria Khalili
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Xinyu Wang
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Jae-Young Cho
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Valerie L Sim
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Marianna Kulka
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada.
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
3
|
Ghosh S, Jana R, Jana S, Basu R, Chatterjee M, Ranawat N, Das Sarma J. Differential expression of cellular prion protein (PrP C) in mouse hepatitis virus induced neuroinflammation. J Neurovirol 2024; 30:215-228. [PMID: 38922550 DOI: 10.1007/s13365-024-01215-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024]
Abstract
The cellular prion protein (PrPC) is an extracellular cell membrane protein. Due to its diversified roles, a definite role of PrPC has been difficult to establish. During viral infection, PrPC has been reported to play a pleiotropic role. Here, we have attempted to envision the function of PrPC in the neurotropic m-CoV-MHV-RSA59-induced model of neuroinflammation in C57BL/6 mice. A significant upregulation of PrPC at protein and mRNA levels was evident in infected mouse brains during the acute phase of neuroinflammation. Furthermore, investigation of the effect of MHV-RSA59 infection on PrPC expression in specific neuronal, microglial, and astrocytoma cell lines, revealed a differential expression of prion protein during neuroinflammation. Additionally, siRNA-mediated downregulation of prnp transcripts reduced the expression of viral antigen and viral infectivity in these cell lines. Cumulatively, our results suggest that PrPC expression significantly increases during acute MHV-RSA59 infection and that PrPC also assists in viral infectivity and viral replication.
Collapse
Affiliation(s)
- Satavisha Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
| | - Rishika Jana
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
| | - Soumen Jana
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
- Optical NeuroImaging Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Rahul Basu
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Madhurima Chatterjee
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
| | - Nishtha Ranawat
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India
- Burke Neurological Institute, Weill Cornell Medicine, New York, NY, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, Mohanpur, 741246, India.
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
4
|
Grimaldi I, Leser FS, Janeiro JM, da Rosa BG, Campanelli AC, Romão L, Lima FRS. The multiple functions of PrP C in physiological, cancer, and neurodegenerative contexts. J Mol Med (Berl) 2022; 100:1405-1425. [PMID: 36056255 DOI: 10.1007/s00109-022-02245-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022]
Abstract
Cellular prion protein (PrPC) is a highly conserved glycoprotein, present both anchored in the cell membrane and soluble in the extracellular medium. It has a diversity of ligands and is variably expressed in numerous tissues and cell subtypes, most notably in the central nervous system (CNS). Its importance has been brought to light over the years both under physiological conditions, such as embryogenesis and immune system homeostasis, and in pathologies, such as cancer and neurodegenerative diseases. During development, PrPC plays an important role in CNS, participating in axonal growth and guidance and differentiation of glial cells, but also in other organs such as the heart, lung, and digestive system. In diseases, PrPC has been related to several types of tumors, modulating cancer stem cells, enhancing malignant properties, and inducing drug resistance. Also, in non-neoplastic diseases, such as Alzheimer's and Parkinson's diseases, PrPC seems to alter the dynamics of neurotoxic aggregate formation and, consequently, the progression of the disease. In this review, we explore in detail the multiple functions of this protein, which proved to be relevant for understanding the dynamics of organism homeostasis, as well as a promising target in the treatment of both neoplastic and degenerative diseases.
Collapse
Affiliation(s)
- Izabella Grimaldi
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Felipe Saceanu Leser
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - José Marcos Janeiro
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bárbara Gomes da Rosa
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Clara Campanelli
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luciana Romão
- Cell Morphogenesis Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Flavia Regina Souza Lima
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
5
|
Extracellular alpha-synuclein: Sensors, receptors, and responses. Neurobiol Dis 2022; 168:105696. [DOI: 10.1016/j.nbd.2022.105696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/28/2022] [Accepted: 03/15/2022] [Indexed: 11/19/2022] Open
|
6
|
Postmortem evidence of brain inflammatory markers in bipolar disorder: a systematic review. Mol Psychiatry 2020; 25:94-113. [PMID: 31249382 DOI: 10.1038/s41380-019-0448-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 03/08/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022]
Abstract
Bipolar disorder (BD) is a chronic affective disorder with extreme mood swings that include mania or hypomania and depression. Though the exact mechanism of BD is unknown, neuroinflammation is one of the numerous investigated etiopathophysiological causes of BD. This article presents a systematic review of the data regarding brain inflammation evaluating microglia, astrocytes, cytokines, chemokines, adhesion molecules, and other inflammatory markers in postmortem BD brain samples. This systematic review was performed according to PRISMA recommendations, and relevant studies were identified by searching the PubMed/MEDLINE, PsycINFO, EMBASE, LILACS, IBECS, and Web of Science databases for peer-reviewed journal articles published by March 2019. Quality of included studies appraised using the QUADAS-2 tool. Among the 1814 articles included in the primary screening, 51 articles measured inflammatory markers in postmortem BD brain samples. A number of studies have shown evidence of inflammation in BD postmortem brain samples. However, an absolute statement cannot be concluded whether neuroinflammation is present in BD due to the large number of studies did not evaluate the presence of infiltrating peripheral immune cells in the central nervous system (CNS) parenchyma, cytokines levels, and microglia activation in the same postmortem brain sample. For example, out of 15 studies that evaluated microglia cells markers, 8 studies found no effect of BD on these cells. Similarly, 17 out of 51 studies evaluating astrocytes markers, 9 studies did not find any effect of BD on astrocyte cells, whereas 8 studies found a decrease and 2 studies presented both increase and decrease in different brain regions. In addition, multiple factors account for the variability across the studies, including postmortem interval, brain area studied, age at diagnosis, undergoing treatment, and others. Future analyses should rectify these potential sources of heterogeneity and reach a consensus regarding the inflammatory markers in postmortem BD brain samples.
Collapse
|
7
|
Abstract
Mammalian prion diseases are a group of neurodegenerative conditions caused by infection of the central nervous system with proteinaceous agents called prions, including sporadic, variant, and iatrogenic Creutzfeldt-Jakob disease; kuru; inherited prion disease; sheep scrapie; bovine spongiform encephalopathy; and chronic wasting disease. Prions are composed of misfolded and multimeric forms of the normal cellular prion protein (PrP). Prion diseases require host expression of the prion protein gene (PRNP) and a range of other cellular functions to support their propagation and toxicity. Inherited forms of prion disease are caused by mutation of PRNP, whereas acquired and sporadically occurring mammalian prion diseases are controlled by powerful genetic risk and modifying factors. Whereas some PrP amino acid variants cause the disease, others confer protection, dramatically altered incubation times, or changes in the clinical phenotype. Multiple mechanisms, including interference with homotypic protein interactions and the selection of the permissible prion strains in a host, play a role. Several non-PRNP factors have now been uncovered that provide insights into pathways of disease susceptibility or neurotoxicity.
Collapse
Affiliation(s)
- Simon Mead
- Medical Research Council Prion Unit at UCL, Institute of Prion Diseases, University College London, London W1W 7FF, United Kingdom;
| | - Sarah Lloyd
- Medical Research Council Prion Unit at UCL, Institute of Prion Diseases, University College London, London W1W 7FF, United Kingdom;
| | - John Collinge
- Medical Research Council Prion Unit at UCL, Institute of Prion Diseases, University College London, London W1W 7FF, United Kingdom;
| |
Collapse
|
8
|
Salvesen Ø, Tatzelt J, Tranulis MA. The prion protein in neuroimmune crosstalk. Neurochem Int 2018; 130:104335. [PMID: 30448564 DOI: 10.1016/j.neuint.2018.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/04/2018] [Accepted: 11/14/2018] [Indexed: 01/11/2023]
Abstract
The cellular prion protein (PrPC) is a medium-sized glycoprotein, attached to the cell surface by a glycosylphosphatidylinositol anchor. PrPC is encoded by a single-copy gene, PRNP, which is abundantly expressed in the central nervous system and at lower levels in non-neuronal cells, including those of the immune system. Evidence from experimental knockout of PRNP in rodents, goats, and cattle and the occurrence of a nonsense mutation in goat that prevents synthesis of PrPC, have shown that the molecule is non-essential for life. Indeed, no easily recognizable phenotypes are associate with a lack of PrPC, except the potentially advantageous trait that animals without PrPC cannot develop prion disease. This is because, in prion diseases, PrPC converts to a pathogenic "scrapie" conformer, PrPSc, which aggregates and eventually induces neurodegeneration. In addition, endogenous neuronal PrPC serves as a toxic receptor to mediate prion-induced neurotoxicity. Thus, PrPC is an interesting target for treatment of prion diseases. Although loss of PrPC has no discernable effect, alteration of its normal physiological function can have very harmful consequences. It is therefore important to understand cellular processes involving PrPC, and research of this topic has advanced considerably in the past decade. Here, we summarize data that indicate the role of PrPC in modulating immune signaling, with emphasis on neuroimmune crosstalk both under basal conditions and during inflammatory stress.
Collapse
Affiliation(s)
- Øyvind Salvesen
- Faculty of Veterinary Medicine, Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Sandnes, Norway.
| | - Jörg Tatzelt
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Germany.
| | - Michael A Tranulis
- Faculty of Veterinary Medicine, Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| |
Collapse
|
9
|
Brás IC, Lopes LV, Outeiro TF. Sensing α-Synuclein From the Outside via the Prion Protein: Implications for Neurodegeneration. Mov Disord 2018; 33:1675-1684. [DOI: 10.1002/mds.27478] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/30/2022] Open
Affiliation(s)
- Inês Caldeira Brás
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration; University Medical Center Göttingen; Göttingen Germany
| | - Luísa V. Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina; Universidade de Lisboa; Lisboa Portugal
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration; University Medical Center Göttingen; Göttingen Germany
- CEDOC, Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciências Médicas; Universidade Nova de Lisboa, Campo dos Mártires da Pátria; Lisboa Portugal
- Max Planck Institute for Experimental Medicine; Göttingen Germany
- Institute of Neuroscience, The Medical School; Newcastle University; Newcastle Upon Tyne UK
| |
Collapse
|
10
|
Megra BW, Eugenin EA, Berman JW. Inflammatory mediators reduce surface PrP c on human BMVEC resulting in decreased barrier integrity. J Transl Med 2018; 98:1347-1359. [PMID: 29959417 PMCID: PMC6163073 DOI: 10.1038/s41374-018-0090-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 05/03/2018] [Accepted: 05/05/2018] [Indexed: 12/18/2022] Open
Abstract
The cellular prion protein (PrPc) is a surface adhesion molecule expressed at junctions of various cell types including brain microvascular endothelial cells (BMVEC) that are important components of the blood-brain barrier (BBB). PrPc is involved in several physiological processes including regulation of epithelial cell barrier function and monocyte migration across BMVEC. BBB dysfunction and disruption are significant events in central nervous system (CNS) inflammatory processes including HIV neuropathogenesis. Tumor necrosis factor (TNF)-α and vascular endothelial growth factor (VEGF) are two inflammatory factors that have been implicated in the processes that affect BBB integrity. To examine the effect of inflammation on PrPc expression in BMVEC, we used these mediators and found that TNF-α and VEGF decrease surface PrPc on primary human BMVEC. We also showed that these factors decrease total PrPc protein as well as mRNA, indicating that they regulate expression of this protein by de novo synthesis. To determine the effect of PrPc loss from the surface of BMVEC on barrier integrity, we used small hairpin RNAs to knockdown PrPc. We found that the absence of PrPc from BMVEC causes increased permeability as determined by a fluorescein isothiocyanate (FITC)-dextran permeability assay. This suggests that cell surface PrPc is essential for endothelial monolayer integrity. To determine the mechanism by which PrPc downregulation leads to increased permeability of an endothelial monolayer, we examined changes in expression and localization of tight junction proteins, occludin and claudin-5, and found that decreased PrPc leads to decreased total and membrane-associated occludin and claudin-5. We propose that an additional mechanism by which inflammatory factors affect endothelial monolayer permeability is by decreasing cell-associated PrPc. This increase in permeability may have subsequent consequences that lead to CNS damage.
Collapse
Affiliation(s)
- Bezawit W. Megra
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Eliseo A. Eugenin
- Public Health Research Institute (PHRI), Newark, NJ 07103,Department of Microbiology and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ 07103
| | - Joan W. Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
11
|
The function of the cellular prion protein in health and disease. Acta Neuropathol 2018; 135:159-178. [PMID: 29151170 DOI: 10.1007/s00401-017-1790-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/11/2022]
Abstract
The essential role of the cellular prion protein (PrPC) in prion disorders such as Creutzfeldt-Jakob disease is well documented. Moreover, evidence is accumulating that PrPC may act as a receptor for protein aggregates and transduce neurotoxic signals in more common neurodegenerative disorders, such as Alzheimer's disease. Although the pathological roles of PrPC have been thoroughly characterized, a general consensus on its physiological function within the brain has not yet been established. Knockout studies in various organisms, ranging from zebrafish to mice, have implicated PrPC in a diverse range of nervous system-related activities that include a key role in the maintenance of peripheral nerve myelination as well as a general ability to protect against neurotoxic stimuli. Thus, the function of PrPC may be multifaceted, with different cell types taking advantage of unique aspects of its biology. Deciphering the cellular function(s) of PrPC and the consequences of its absence is not simply an academic curiosity, since lowering PrPC levels in the brain is predicted to be a powerful therapeutic strategy for the treatment of prion disease. In this review, we outline the various approaches that have been employed in an effort to uncover the physiological and pathological functions of PrPC. While these studies have revealed important clues about the biology of the prion protein, the precise reason for PrPC's existence remains enigmatic.
Collapse
|
12
|
Knock E, Matsuzaki S, Takamura H, Satoh K, Rooke G, Han K, Zhang H, Staniszewski A, Katayama T, Arancio O, Fraser PE. SUMO1 impact on Alzheimer disease pathology in an amyloid-depositing mouse model. Neurobiol Dis 2018; 110:154-165. [DOI: 10.1016/j.nbd.2017.11.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 11/20/2017] [Accepted: 11/29/2017] [Indexed: 12/27/2022] Open
|
13
|
Beckman D, Linden R. A roadmap for investigating the role of the prion protein in depression associated with neurodegenerative disease. Prion 2017; 10:131-42. [PMID: 27057694 DOI: 10.1080/19336896.2016.1152437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The physiological properties of the native, endogenous prion protein (PrP(C)) is a matter of concern, due to its pleiotropic functions and links to neurodegenerative disorders and cancer. In line with our hypothesis that the basic function of PrP(C) is to serve as a cell surface scaffold for the assembly of signaling modules, multiple interactions have been identified of PrP(C) with signaling molecules, including neurotransmitter receptors. We recently reported evidence that PrP(C) may modulate monoaminergic neurotransmission, as well as depressive-like behavior in mice. Here, we discuss how those results, together with a number of other studies, including our previous demonstration that both inflammatory and behavioral stress modulate PrP(C) content in neutrophils, suggest a distributed role of PrP(C) in clinical depression and inflammation associated with neurodegenerative diseases. An overarching understanding of the multiple interventions of PrP(C) upon physiological events may both shed light on the pathogenesis of, as well as help the identification of novel therapeutic targets for clinical depression, Prion and Alzheimer's Diseases.
Collapse
Affiliation(s)
| | - Rafael Linden
- a Instituto de Biofísica da UFRJ, Rio de Janeiro , Brazil
| |
Collapse
|
14
|
Bakkebø MK, Mouillet-Richard S, Espenes A, Goldmann W, Tatzelt J, Tranulis MA. The Cellular Prion Protein: A Player in Immunological Quiescence. Front Immunol 2015; 6:450. [PMID: 26388873 PMCID: PMC4557099 DOI: 10.3389/fimmu.2015.00450] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/19/2015] [Indexed: 01/09/2023] Open
Abstract
Despite intensive studies since the 1990s, the physiological role of the cellular prion protein (PrP(C)) remains elusive. Here, we present a novel concept suggesting that PrP(C) contributes to immunological quiescence in addition to cell protection. PrP(C) is highly expressed in diverse organs that by multiple means are particularly protected from inflammation, such as the brain, eye, placenta, pregnant uterus, and testes, while at the same time it is expressed in most cells of the lymphoreticular system. In this paradigm, PrP(C) serves two principal roles: to modulate the inflammatory potential of immune cells and to protect vulnerable parenchymal cells against noxious insults generated through inflammation. Here, we review studies of PrP(C) physiology in view of this concept.
Collapse
Affiliation(s)
- Maren K. Bakkebø
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | | | - Arild Espenes
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Wilfred Goldmann
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Jörg Tatzelt
- Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Michael A. Tranulis
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway,*Correspondence: Michael A. Tranulis, Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Campus Adamstuen, Oslo 0033, Norway,
| |
Collapse
|
15
|
Activation and function of murine primary microglia in the absence of the prion protein. J Neuroimmunol 2015; 286:25-32. [PMID: 26298321 DOI: 10.1016/j.jneuroim.2015.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/02/2015] [Accepted: 07/04/2015] [Indexed: 11/21/2022]
Abstract
The prion protein (PrP(C)) is predominantly expressed in the nervous and immune systems and is involved in relevant cell signaling. Microglia participate in neuroimmune interactions, and their regulatory mechanisms are critical for both health and disease. Despite recent reports with a microglial cell line, little is known about the relevance of PrP(C) in brain microglia. We investigated the role of PrP(C) in mouse primary microglia, and found no differences between wild type and Prnp-null cells in cell morphology or the expression of a microglial marker. Translocation of NF-κB to the nucleus also did not differ, nor did cytokine production. The levels of iNOS were also similar and, finally, microglia of either genotype showed no differences in either rates of phagocytosis or migration, even following activation. Thus, functional roles of PrP(C) in primary microglial cells are - if present - much more subtle than in transformed microglial cell lines.
Collapse
|
16
|
Urso E, Manno D, Serra A, Buccolieri A, Rizzello A, Danieli A, Acierno R, Salvato B, Maffia M. Role of the cellular prion protein in the neuron adaptation strategy to copper deficiency. Cell Mol Neurobiol 2012; 32:989-1001. [PMID: 22362149 PMCID: PMC11498592 DOI: 10.1007/s10571-012-9815-5] [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: 12/22/2011] [Accepted: 02/07/2012] [Indexed: 01/15/2023]
Abstract
Copper transporter 1 (CTR1), cellular prion protein (PrP(C)), natural resistance-associated macrophage protein 2 (NRAMP2) and ATP7A proteins control the cell absorption and efflux of copper (Cu) ions in nervous tissues upon physiological conditions. Little is known about their regulation under reduced Cu availability, a condition underlying the onset of diffused neurodegenerative disorders. In this study, rat neuron-like cells were exposed to Cu starvation for 48 h. The activation of Caspase-3 enzymes and the impairment of Cu,Zn superoxide dismutase (Cu,Zn SOD) activity depicted the initiation of a pro-apoptotic program, preliminary to the appearance of the morphological signs of apoptosis. The transcriptional response related to Cu transport proteins has been investigated. Notably, PrP(C) transcript and protein levels were consistently elevated upon Cu deficiency. The CTR1 protein amount was stable, despite a two-fold increase in the transcript amount, meaning the activation of post-translational regulatory mechanisms. NRAMP2 and ATP7A expressions were unvaried. The up-regulated PrP(C) has been demonstrated to enhance the cell Cu uptake ability by about 50% with respect to the basal transport, and so sustain the Cu delivery to the Cu,Zn SOD cuproenzymes. Conclusively, the study suggests a pivotal role for PrP(C) in the cell adaptation to Cu limitation through a direct activity of ion uptake. In this view, the PrP(C) accumulation observed in several cancer cell lines could be interpreted as a molecular marker of cell Cu deficiency and a potential target of therapeutic interventions against disorders caused by metal imbalances.
Collapse
Affiliation(s)
- Emanuela Urso
- Department of Biological and Environmental Sciences and Technologies, University of Salento, SP Lecce-Monteroni, 73100 Lecce, Italy
| | - Daniela Manno
- Department of Material Sciences, University of Salento, SP Lecce-Monteroni, 73100 Lecce, Italy
| | - Antonio Serra
- Department of Material Sciences, University of Salento, SP Lecce-Monteroni, 73100 Lecce, Italy
| | - Alessandro Buccolieri
- Department of Material Sciences, University of Salento, SP Lecce-Monteroni, 73100 Lecce, Italy
| | - Antonia Rizzello
- Department of Biological and Environmental Sciences and Technologies, University of Salento, SP Lecce-Monteroni, 73100 Lecce, Italy
| | - Antonio Danieli
- Department of Biological and Environmental Sciences and Technologies, University of Salento, SP Lecce-Monteroni, 73100 Lecce, Italy
| | - Raffaele Acierno
- Department of Biological and Environmental Sciences and Technologies, University of Salento, SP Lecce-Monteroni, 73100 Lecce, Italy
| | - Benedetto Salvato
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35121 Padua, Italy
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, SP Lecce-Monteroni, 73100 Lecce, Italy
| |
Collapse
|
17
|
Llorens F, Del Río JA. Unraveling the neuroprotective mechanisms of PrP (C) in excitotoxicity. Prion 2012; 6:245-51. [PMID: 22437735 DOI: 10.4161/pri.19639] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Knowledge of the natural roles of cellular prion protein (PrP (C) ) is essential to an understanding of the molecular basis of prion pathologies. This GPI-anchored protein has been described in synaptic contacts, and loss of its synaptic function in complex systems may contribute to the synaptic loss and neuronal degeneration observed in prionopathy. In addition, Prnp knockout mice show enhanced susceptibility to several excitotoxic insults, GABAA receptor-mediated fast inhibition was weakened, LTP was modified and cellular stress increased. Although little is known about how PrP (C) exerts its function at the synapse or the downstream events leading to PrP (C) -mediated neuroprotection against excitotoxic insults, PrP (C) has recently been reported to interact with two glutamate receptor subunits (NR2D and GluR6/7). In both cases the presence of PrP (C) blocks the neurotoxicity induced by NMDA and Kainate respectively. Furthermore, signals for seizure and neuronal cell death in response to Kainate in Prnp knockout mouse are associated with JNK3 activity, through enhancing the interaction of GluR6 with PSD-95. In combination with previous data, these results shed light on the molecular mechanisms behind the role of PrP (C) in excitotoxicity. Future experimental approaches are suggested and discussed.
Collapse
Affiliation(s)
- Franc Llorens
- Molecular and Cellular Neurobiotechnology Group, Institut de Bioenginyeria de Catalunya (IBEC), Parc Científic de Barcelona, Barcelona, Spain.
| | | |
Collapse
|
18
|
Bribián A, Fontana X, Llorens F, Gavín R, Reina M, García-Verdugo JM, Torres JM, de Castro F, del Río JA. Role of the cellular prion protein in oligodendrocyte precursor cell proliferation and differentiation in the developing and adult mouse CNS. PLoS One 2012; 7:e33872. [PMID: 22529900 PMCID: PMC3329524 DOI: 10.1371/journal.pone.0033872] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 02/18/2012] [Indexed: 11/18/2022] Open
Abstract
There are numerous studies describing the signaling mechanisms that mediate oligodendrocyte precursor cell (OPC) proliferation and differentiation, although the contribution of the cellular prion protein (PrPc) to this process remains unclear. PrPc is a glycosyl-phosphatidylinositol (GPI)-anchored glycoprotein involved in diverse cellular processes during the development and maturation of the mammalian central nervous system (CNS). Here we describe how PrPc influences oligodendrocyte proliferation in the developing and adult CNS. OPCs that lack PrPc proliferate more vigorously at the expense of a delay in differentiation, which correlates with changes in the expression of oligodendrocyte lineage markers. In addition, numerous NG2-positive cells were observed in cortical regions of adult PrPc knockout mice, although no significant changes in myelination can be seen, probably due to the death of surplus cells.
Collapse
Affiliation(s)
- Ana Bribián
- Molecular and Cellular Neurobiotechnology, Catalonian Institute for Bioengineering (IBEC), Parc Científic de Barcelona, Barcelona, Spain
- Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Xavier Fontana
- Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain
| | - Franc Llorens
- Molecular and Cellular Neurobiotechnology, Catalonian Institute for Bioengineering (IBEC), Parc Científic de Barcelona, Barcelona, Spain
- Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Rosalina Gavín
- Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Manuel Reina
- Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain
| | - José Manuel García-Verdugo
- Laboratorio de Neurobiología Comparada, Instituto Cabanillas de Biodiversidad y Biología Evolutiva, Universidad de Valencia, Valencia, Spain
| | - Juan María Torres
- Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Spain
| | - Fernando de Castro
- GNDe-Grupo de Neurobiología del Desarrollo, Unidad de Neurología Experimental, Hospital Nacional de Parapléjicos, Toledo, Spain
- Instituto Cajal-CSIC, Madrid, Spain
| | - José Antonio del Río
- Molecular and Cellular Neurobiotechnology, Catalonian Institute for Bioengineering (IBEC), Parc Científic de Barcelona, Barcelona, Spain
- Department of Cell Biology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- * E-mail:
| |
Collapse
|
19
|
da Fonseca A, Romão L, Amaral R, Assad Kahn S, Lobo D, Martins S, Marcondes de Souza J, Moura-Neto V, Lima F. Microglial stress inducible protein 1 promotes proliferation and migration in human glioblastoma cells. Neuroscience 2012; 200:130-41. [DOI: 10.1016/j.neuroscience.2011.10.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 10/10/2011] [Accepted: 10/14/2011] [Indexed: 12/26/2022]
|
20
|
The cellular prion protein in multiple sclerosis: A potential target for neurotherapeutics? Transl Neurosci 2011. [DOI: 10.2478/s13380-011-0042-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractMultiple sclerosis (MS) is a debilitating disease that affects millions. There is no known cure for the disease and neither is the cause of the disease known. Recent studies have indicated that it is a multi-factorial disease with several genes involved. Importantly, sunlight and vitamin D have been implicated in the progression of the disease. The pathogenesis of MS chiefly involves loss of oligodendrocytes, which in addition to being killed by inflammatory mediators in the CNS, also succumbs to loss of trophic support from astrocytes. Neurotrophins play an important role in myelination and the cellular prion protein (PrPC) is a key player in this process. Although the physiological roles of PrPC remain to be fully understood, increasing evidence suggests multiple roles for PrPC in regulation of cellular immunity and for its interaction with several neurotrophins that are necessary for homeostasis of the nervous system. This mini-review focuses on the findings establishing a crucial role for PrPC in the neuropathogenesis of MS, emphasizing its neuroprotective role. Since MS is a multi-factorial disease with unknown etiology and no cure, this review aims to highlight endogenous repair mechanisms mediated by PrPC that might contribute to functional recovery in MS patients.
Collapse
|
21
|
Alier K, Li Z, Mactavish D, Westaway D, Jhamandas JH. Ionic mechanisms of action of prion protein fragment PrP(106-126) in rat basal forebrain neurons. J Neurosci Res 2010; 88:2217-27. [PMID: 20175205 DOI: 10.1002/jnr.22372] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Prion diseases are neurodegenerative disorders that are characterized by the presence of the misfolded prion protein (PrP). Neurotoxicity in these diseases may result from prion-induced modulation of ion channel function, changes in neuronal excitability, and consequent disruption of cellular homeostasis. We therefore examined PrP effects on a suite of potassium (K(+)) conductances that govern excitability of basal forebrain neurons. Our study examined the effects of a PrP fragment [PrP(106-126), 50 nM] on rat neurons using the patch clamp technique. In this paradigm, PrP(106-126) peptide, but not the "scrambled" sequence of PrP(106-126), evoked a reduction of whole-cell outward currents in a voltage range between -30 and +30 mV. Reduction of whole-cell outward currents was significantly attenuated in Ca(2+)-free external media and also in the presence of iberiotoxin, a blocker of calcium-activated potassium conductance. PrP(106-126) application also evoked a depression of the delayed rectifier (I(K)) and transient outward (I(A)) potassium currents. By using single cell RT-PCR, we identified the presence of two neuronal chemical phenotypes, GABAergic and cholinergic, in cells from which we recorded. Furthermore, cholinergic and GABAergic neurons were shown to express K(v)4.2 channels. Our data establish that the central region of PrP, defined by the PrP(106-126) peptide used at nanomolar concentrations, induces a reduction of specific K(+) channel conductances in basal forebrain neurons. These findings suggest novel links between PrP signalling partners inferred from genetic experiments, K(+) channels, and PrP-mediated neurotoxicity.
Collapse
Affiliation(s)
- Kwai Alier
- Department of Medicine (Neurology), University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
22
|
Roberts TK, Eugenin EA, Morgello S, Clements JE, Zink MC, Berman JW. PrPC, the cellular isoform of the human prion protein, is a novel biomarker of HIV-associated neurocognitive impairment and mediates neuroinflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1848-60. [PMID: 20724601 DOI: 10.2353/ajpath.2010.091006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Of the 33 million people infected with the human immunodeficiency virus (HIV) worldwide, 40-60% of individuals will eventually develop neurocognitive sequelae that can be attributed to the presence of HIV-1 in the central nervous system (CNS) and its associated neuroinflammation despite antiretroviral therapy. PrP(C) (protease resistant protein, cellular isoform) is the nonpathological cellular isoform of the human prion protein that participates in many physiological processes that are disrupted during HIV-1 infection. However, its role in HIV-1 CNS disease is unknown. We demonstrate that PrP(C) is significantly increased in both the CNS of HIV-1-infected individuals with neurocognitive impairment and in SIV-infected macaques with encephalitis. PrP(C) is released into the cerebrospinal fluid, and its levels correlate with CNS compromise, suggesting it is a biomarker of HIV-associated neurocognitive impairment. We show that the chemokine (c-c Motif) Ligand-2 (CCL2) increases PrP(C) release from CNS cells, while HIV-1 infection alters PrP(C) release from peripheral blood mononuclear cells. Soluble PrP(C) mediates neuroinflammation by inducing astrocyte production of both CCL2 and interleukin 6. This report presents the first evidence that PrP(C) dysregulation occurs in cognitively impaired HIV-1-infected individuals and that PrP(C) participates in the pathogenesis of HIV-1-associated CNS disease.
Collapse
Affiliation(s)
- Toni K Roberts
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | |
Collapse
|
23
|
Urso E, Rizzello A, Acierno R, Lionetto MG, Salvato B, Storelli C, Maffia M. Fluorimetric Analysis of Copper Transport Mechanisms in the B104 Neuroblastoma Cell Model: A Contribution from Cellular Prion Protein to Copper Supplying. J Membr Biol 2009; 233:13-21. [DOI: 10.1007/s00232-009-9219-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2009] [Accepted: 11/05/2009] [Indexed: 12/15/2022]
|
24
|
Zhang P, Wong TA, Lokuta KM, Turner DE, Vujisic K, Liu B. Microglia enhance manganese chloride-induced dopaminergic neurodegeneration: role of free radical generation. Exp Neurol 2009; 217:219-30. [PMID: 19268665 DOI: 10.1016/j.expneurol.2009.02.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Revised: 02/16/2009] [Accepted: 02/19/2009] [Indexed: 11/15/2022]
Abstract
Exposure to elevated levels of manganese has been shown to cause neuronal damage in the midbrain and the development of Parkinsonian symptoms. Activation of microglia and release of neurotoxic factors in particular free radicals are known to contribute to neurodegeneration. We have recently reported that manganese chloride (MnCl(2)) stimulates microglia to produce reactive oxygen species (ROS). The aim of this study is to determine the role of microglia in the MnCl(2)-induced degeneration of dopaminergic (DA) neurons that are particularly vulnerable to oxidative insult. MnCl(2) (10-300 microM; 7 days) was markedly more effective in damaging DA neurons in the rat mesencephalic neuron-glia cultures than the neuron-enriched (microglia-depleted) cultures. In addition, the microglia-enhanced MnCl(2) toxicity was found to be preferential to DA neurons. The microglial enhancement of DA neurotoxicity was further supported by the observation that replenishment of microglia to the neuron-enriched cultures significantly increased the susceptibility of DA neurons to the MnCl(2)-induced damage. Analysis of the temporal relationship between microglial activation and DA neurodegeneration revealed that MnCl(2)-stimulated microglial activation preceded DA neurodegeneration. Mechanistically, MnCl(2) (10-300 microM) stimulated a concentration- and time-dependent robust production of ROS and moderate production of nitric oxide but no detectable release of tumor necrosis factor-alpha and interleukin-1beta. Application of free radical scavengers including superoxide dismutase/catalase, glutathione, N-acetyl cysteine and an inhibitor of nitric oxide biosynthesis significantly protected DA neurons against the MnCl(2)-induced degeneration. These results demonstrate that microglial activation and the production of reactive nitrogen and oxygen free radicals promote the MnCl(2)-induced DA neurodegeneration.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | |
Collapse
|
25
|
Tsutsui S, Hahn JN, Johnson TA, Ali Z, Jirik FR. Absence of the cellular prion protein exacerbates and prolongs neuroinflammation in experimental autoimmune encephalomyelitis. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:1029-41. [PMID: 18815152 DOI: 10.2353/ajpath.2008.071062] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although the physiological roles of the cellular prion protein (PrP C) remain to be fully elucidated, PrP C has been proposed to represent a potential regulator of cellular immunity. To test this hypothesis, we evaluated the consequences of PrP C deficiency on the course of experimental autoimmune encephalomyelitis induced by immunization with myelin oligodendrocyte glycoprotein peptide. Consistent with augmented proliferative responses and increased cytokine gene expression by myelin oligodendrocyte glycoprotein-primed Prnp-/- T cells, PrP C-deficient mice demonstrated more aggressive disease onset and a lack of clinical improvement during the chronic phase of experimental autoimmune encephalomyelitis. Acutely, Prnp-/- spinal cord, cerebellum, and forebrain exhibited higher levels of leukocytic infiltrates and pro-inflammatory cytokine gene expression, as well as increased spinal cord myelin basic protein and axonal loss. During the chronic phase, a remarkable persistence of leukocytic infiltrates was present in the forebrain and cerebellum, accompanied by an increase in interferon-gamma and interleukin-17 transcripts. Attenuation of T cell-dependent neuroinflammation thus represents a potential novel function of PrP C.
Collapse
Affiliation(s)
- Shigeki Tsutsui
- Department of Biochemistry and Molecular Biology, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
26
|
The polybasic N-terminal region of the prion protein controls the physical properties of both the cellular and fibrillar forms of PrP. J Mol Biol 2008; 383:1210-24. [PMID: 18789949 DOI: 10.1016/j.jmb.2008.08.073] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 07/23/2008] [Accepted: 08/25/2008] [Indexed: 01/07/2023]
Abstract
Individual variations in structure and morphology of amyloid fibrils produced from a single polypeptide are likely to underlie the molecular origin of prion strains and control the efficiency of the species barrier in the transmission of prions. Previously, we observed that the shape of amyloid fibrils produced from full-length prion protein (PrP 23-231) varied substantially for different batches of purified recombinant PrP. Variations in fibril morphology were also observed for different fractions that corresponded to the highly pure PrP peak collected at the last step of purification. A series of biochemical experiments revealed that the variation in fibril morphology was attributable to the presence of miniscule amounts of N-terminally truncated PrPs, where a PrP encompassing residue 31-231 was the most abundant of the truncated polypeptides. Subsequent experiments showed that the presence of small amounts of recombinant PrP 31-231 (0.1-1%) in mixtures with full-length PrP 23-231 had a dramatic impact on fibril morphology and conformation. Furthermore, the deletion of the short polybasic N-terminal region 23-30 was found to reduce the folding efficiency to the native alpha-helical forms and the conformational stability of alpha-PrP. These findings are very surprising considering that residues 23-30 are very distant from the C-terminal globular folded domain in alpha-PrP and from the prion folding domain in the fibrillar form. However, our studies suggest that the N-terminal polybasic region 23-30 is essential for effective folding of PrP to its native cellular conformation. This work also suggests that this region could regulate diversity of prion strains or subtypes despite its remote location from the prion folding domain.
Collapse
|
27
|
Isaacs JD, Garden OA, Kaur G, Collinge J, Jackson GS, Altmann DM. The cellular prion protein is preferentially expressed by CD4+ CD25+ Foxp3+ regulatory T cells. Immunology 2008; 125:313-9. [PMID: 18462346 DOI: 10.1111/j.1365-2567.2008.02853.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Post-translational modification of the cellular prion protein (PrP(C)) is intimately associated with the pathogenesis of prion disease, yet the normal function of the protein remains unclear. PrP(C) is expressed in lymphoid cells and is known to be a T-cell activation antigen. Further, transcription profiling studies of regulatory T cells have shown preferential overexpression of PrP(C), suggesting a possible role in regulatory function. We report that both the expression of PrP message and cell surface PrP(C) levels are increased in murine CD4(+) CD25(+) regulatory T cells compared with CD4(+) CD25(-) cells. However, PrP(0/0) mice do not show altered regulatory T-cell numbers or forkhead box P3 (Foxp3) expression levels, or impaired regulatory T-cell function in vitro. Nevertheless, the preferential expression of surface PrP(C) by regulatory T cells raises the possibility that therapeutic ligation of PrP(C) might alter immune regulation.
Collapse
Affiliation(s)
- Jeremy D Isaacs
- Department of Infectious Diseases and Immunity, Imperial College, Hammersmith Hospital, London, UK
| | | | | | | | | | | |
Collapse
|
28
|
Effects of nutrition and genotype on prion protein (PrPC) gene expression in the fetal and maternal sheep placenta. Placenta 2008; 29:422-8. [PMID: 18358531 DOI: 10.1016/j.placenta.2008.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 01/24/2008] [Accepted: 02/09/2008] [Indexed: 11/21/2022]
Abstract
For placental transmission of scrapie to occur, the normal cellular prion protein (PrPC) must be converted to an abnormal infectious form known as PrPSc. PrPC genotype influences susceptibility to contracting scrapie, but we still do not understand whether genotype or expression levels of PrPC are important in transmission of scrapie. Some evidence exists that nutrition affects expression levels of PrPC. Thus, we evaluated the effects of genotype and nutrition on PrPC mRNA and protein expression in adolescent ewes fed at control (100% of National Research Council [NRC] requirements) or restricted (60% of NRC) levels of diet intake during two periods of pregnancy (days 50-90 and days 90-130)]. Gravid uteri (n=50) from singleton pregnancies were collected at day 130, and placentomes were either separated into caruncular (CAR; maternal) or cotyledonary (COT; fetal) placenta and snap-frozen for PrPC mRNA expression or perfusion fixed for PrPC protein expression. PrPC genotypes were determined (codons 136 and 171) using SNP assay. There were no genotype effects on PrPC mRNA expression in CAR or on PrPC protein expression in either CAR or COT, but PrPC mRNA expression in COT was greater (P<0.02) when codon 136 was homozygous for alanine. Some PrPC protein-positive cells were found in the epithelium of CAR, but most were found in trophoblast binucleate and mononucleate cells of COT. In CAR, from days 90 to 130, PrPC protein abundance was greater (P=0.003) in diet-restricted ewes than in control ewes, but was less uniformly distributed (P<0.007). Additionally, in COT, from days 90 to 130, PrPC protein was less uniformly distributed (P<0.01) in diet-restricted ewes. The localized increase in PrPC protein expression, found in ewes diet-restricted late in pregnancy, may suggest a protective role for PrPC in placental biology. Further study is needed to evaluate whether nutrition, PrPC genotype, and PrPC expression levels influence placental transmission of scrapie.
Collapse
|
29
|
Weis S, Haybaeck J, Dulay JR, Llenos IC. Expression of cellular prion protein (PrPc) in schizophrenia, bipolar disorder, and depression. J Neural Transm (Vienna) 2008; 115:761-71. [DOI: 10.1007/s00702-007-0013-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 11/19/2007] [Indexed: 01/27/2023]
|
30
|
Ning ZY, Zhao DM, Yang JM, Cui YL, Meng LP, Wu CD, Liu HX. QUANTIFICATION OF PRION GENE EXPRESSION IN BRAIN AND PERIPHERAL ORGANS OF GOLDEN HAMSTER BY REAL-TIME RT-PCR. Anim Biotechnol 2007; 16:55-65. [PMID: 15926263 DOI: 10.1081/abio-200053404] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Determination of tissue-specific expression of cellular prion protein (PrPc) is essential for understanding its poorly explained role in organisms. Herein we report on quantification of PrP mRNA in golden hamsters, a popular experimental model for studying mechanisms of transmissible spongiform encephalopathies (TSE), by real-time RT-PCR. Total RNA was isolated from four different regions of the brain and six peripheral organs of eight golden hamsters. PrP mRNA copy numbers were determined using absolute standard curve method with real-time quantitative PCR instrument. It was found that high mRNA levels were present in all four regions of the brain examined, including obex, neocortex, cerebellum, and thalamus. In peripheral organs examined, inguinal lymph node showed high level of the expression similar to that in overall brain; spleen, heart, liver, and lung showed moderate levels of the expression; and kidney showed the lowest expression. Our result is consistent with the potential involvement of different organs in prion diseases and offers essential data for further study of TSE mechanism in this animal model.
Collapse
Affiliation(s)
- Zhang-Yong Ning
- National Animal Transmissible Spongiform Encephalopathies Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | | | | | | | | | | | | |
Collapse
|
31
|
Pasquali P, Nonno R, Mandara MT, Di Bari MA, Ricci G, Petrucci P, Capuccini S, Cartoni C, Macrì A, Agrimi U. Intracerebral administration of interleukin-12 (IL-12) and IL-18 modifies the course of mouse scrapie. BMC Vet Res 2006; 2:37. [PMID: 17192191 PMCID: PMC1769363 DOI: 10.1186/1746-6148-2-37] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Accepted: 12/27/2006] [Indexed: 11/10/2022] Open
Abstract
Background Prion diseases are characterised by a neurodegenerative pattern in which the function of immune system remains still elusive. In the present study, we evaluate if an exogenous treatment with Interleukin-12 (IL-12) and IL-18, able to activate microglia, is able to affect scrapie pathogenesis. Results Cytokines injected intracranially, induced a strong inflammatory response characterised by TNF-α production and microglia activation. Two groups of mice were injected intracerebrally with high dose of ME7 strain of scrapie containing IL-12 and IL-18 or sterile saline. Cytokines-treated mice showed a more pronounced accumulation of PrPSc in brain tissues at 90 days post-inoculation and a shorter mean survival times than untreated mice. Conclusion We can conclude that intracerebral administration of IL-12 and IL-18 can modulate scrapie pathogenesis possibly through a microglia-mediated pattern.
Collapse
Affiliation(s)
- Paolo Pasquali
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Romolo Nonno
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Maria Teresa Mandara
- Department of Biopathological Veterinary Science, Veterinary Medicine School, Università degli Studi di Perugia, Perugia, Italy
| | - Michele Angelo Di Bari
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Giovanni Ricci
- Department of Biopathological Veterinary Science, Veterinary Medicine School, Università degli Studi di Perugia, Perugia, Italy
| | - Paola Petrucci
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Silvia Capuccini
- Department of Biopathological Veterinary Science, Veterinary Medicine School, Università degli Studi di Perugia, Perugia, Italy
| | - Claudia Cartoni
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Agostino Macrì
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Umberto Agrimi
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
32
|
Abstract
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 widely in the immune system, in haematopoietic stem cells and mature lymphoid and myeloid compartments in addition to cells of the central nervous system. It is up-regulated in T cell activation and may be expressed at higher levels by specialized classes of lymphocyte. Furthermore, antibody cross-linking of surface PrP modulates T cell activation and leads to rearrangements of lipid raft constituents and increased phosphorylation of signalling proteins. These findings appear to indicate an important but, as yet, ill-defined role in T cell function. Although PrP(-/-) mice have been reported to have only minor alterations in immune function, recent work has suggested that PrP is required for self-renewal of haematopoietic stem cells. Here, we consider the evidence for a distinctive role for PrP(C) in the immune system and what the effects of anti-prion therapeutics may be on immune function.
Collapse
Affiliation(s)
- J D Isaacs
- Human Disease Immunogenetics Group, Department of Infectious Diseases and Immunity, Imperial College London, Hammersmith Hospital, London, UK
| | | | | |
Collapse
|
33
|
Adle-Biassette H, Verney C, Peoc'h K, Dauge MC, Razavi F, Choudat L, Gressens P, Budka H, Henin D. Immunohistochemical expression of prion protein (PrPC) in the human forebrain during development. J Neuropathol Exp Neurol 2006; 65:698-706. [PMID: 16825956 DOI: 10.1097/01.jnen.0000228137.10531.72] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The cellular prion protein (PrPC) is a ubiquitous protein whose expression in the adult brain occurs mainly in synapses. We used monoclonal antibodies to study fetal and perinatal PrPC expression in the human forebrain. Double immunofluorescence and confocal microscopy with GFAP, Iba1, MAP2, doublecortin, synaptophysin, and GAP-43 were used to localize PrPC. PrPC immunoreactivity was observed in axonal tracts and fascicles from the 11th week to the end of gestation. Synapses expressed PrPC at increasing levels throughout synaptogenesis. At midgestation, a few PrPC-labeled neurons were detected in the cortical anlage and numerous ameboid and intermediate microglial cells were PrPC-positive. In contrast, at the end of gestation, microglial PrPC expression decreased to almost nothing, whereas neuronal PrPC expression increased, most notably in ischemic areas. In adults, PrPC immunoreactivity was restricted to the synaptic neuropil of the gray matter. At all ages, choroid plexus, ependymal, and endothelial cells were labeled, whereas astrocytes were only occasionally immunoreactive. In conclusion, the early expression of PrPC in the axonal field may suggest a specific role for this molecule in axonal growth during development. Moreover, PrPC may play a role in early microglial cell development.
Collapse
Affiliation(s)
- Homa Adle-Biassette
- AP HP, Hôpital Bichat-Claude Bernard, Service d'Anatomie Pathologie, Université Paris 7, Faculté de Médecine Denis Diderot, Paris, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Shyu WC, Lin SZ, Chiang MF, Ding DC, Li KW, Chen SF, Yang HI, Li H. Overexpression of PrPC by adenovirus-mediated gene targeting reduces ischemic injury in a stroke rat model. J Neurosci 2006; 25:8967-77. [PMID: 16192387 PMCID: PMC6725592 DOI: 10.1523/jneurosci.1115-05.2005] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Prion diseases are induced by pathologically misfolded prion protein (PrPSc), which recruit normal sialoglycoprotein PrPC by a template-directed process. In this study, we investigated the expression of PrPC in a rat model of cerebral ischemia to more fully understand its physiological role. Immunohistochemical analysis demonstrated that PrPC-immunoreactive cells increased significantly in the penumbra of ischemic rat brain compared with the untreated brain. Western blot analysis showed that PrPC protein expression increased in ischemic brain tissue in a time-dependent manner. In addition, PrPC protein expression was seen to colocalize with neuron, glial, and vascular endothelial cells in the penumbric region of the ischemic brain. Overexpression of PrPC by injection of rAd (replication-defective recombinant adenoviral)-PGK (phosphoglycerate kinase)-PrPC-Flag into ischemic rat brain improved neurological behavior and reduced the volume of cerebral infarction, which is supportive of a role for PrPC in the neuroprotective adaptive cellular response to ischemic lesions. Concomitant upregulation of PrPC and activated extracellular signal-regulated kinase (ERK1/2) under hypoxia-reoxygenation in primary cortical cultures was shown to be dependent on ERK1/2 phosphorylation. During hypoxia-reoxygenation, mouse neuroblastoma cell line N18 cells transfected with luciferase rat PrPC promoter reporter constructs, containing the heat shock element (HSE), expressed higher luciferase activities (3- to 10-fold) than those cells transfected with constructs not containing HSE. We propose that HSTF-1 (hypoxia-activated transcription factor), phosphorylated by ERK1/2, may in turn interact with HSE in the promoter of PrPC resulting in gene expression of the prion gene. In summary, we conclude that upregulation of PrPC expression after cerebral ischemia and hypoxia exerts a neuroprotective effect on injured neural tissue. This study suggests that PrPC has physiological relevance to cerebral ischemic injury and could be useful as a therapeutic target for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Woei-Cherng Shyu
- Department of Neurology, Neuro-Medical Scientific Center, Tzu-Chi Buddhist General Hospital, Tzu-Chi University, Hualien, 970, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Members of the nuclear factor kappaB (NF-kappaB) family of transcription factors are activated within the CNS in pathological settings of apoptosis and neurological disease. Recent work using several model systems provides accumulating evidence that these transcription factors also participate in the regulation of neuronal activity-dependent transcription and behavior under physiological conditions. This review highlights advances in our understanding of the mechanisms of Ca(2+)-responsive activation and synaptic signaling to the nucleus by NF-kappaB transcription factors within the CNS, and the relevance of this transcription factor family for learning and memory.
Collapse
Affiliation(s)
- Mollie K Meffert
- Division of Biology, MC204-31 California Institute of Technology, Pasadena, CA 91125, USA.
| | | |
Collapse
|
36
|
Liberski PP, Sikorska B, Bratosiewicz-Wasik J, Gajdusek DC, Brown P. Neuronal cell death in transmissible spongiform encephalopathies (prion diseases) revisited: from apoptosis to autophagy. Int J Biochem Cell Biol 2005; 36:2473-90. [PMID: 15325586 DOI: 10.1016/j.biocel.2004.04.016] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Neuronal autophagy, like apoptosis, is one of the mechanisms of the programmed cell death (PCD). In this review, we summarize the presence of autophagic vacuoles in experimentally induced scrapie, Creutzfeldt-Jakob disease and Gerstmann-Sträussler-Scheinker (GSS) syndrome. Initially, a part of the neuronal cytoplasm was sequestrated by concentric arrays of double membranes; the enclosed cytoplasm appeared relatively normal except that its density was often increased. Next, electron density of the central area dramatically increased. The membranes then proliferated within the cytoplasm in a labyrinth-like manner and the area sequestrated by these membranes enlarged into a more complex structure consisting of vacuoles, electron-dense areas and areas of normally-looking cytoplasm connected by convoluted membranes. Of note, autophagic vacuoles form not only in neuronal perikarya but also in neurites and synapses. Finally, a large area of the cytoplasm was transformed into a collection of autophagic vacuoles of different sizes. On a basis of ultrastructural studies, we suggest that autophagy plays a major role in transmissible spongiform encephalopathies (TSEs) and may even participate in a formation of spongiform change.
Collapse
Affiliation(s)
- Pawel P Liberski
- Department of Molecular Pathology and Neuropathology, Medical University Lodz, Czechoslowacka Street 8/10; pl 92-216 Lodz, Poland.
| | | | | | | | | |
Collapse
|
37
|
de Almeida CJG, Chiarini LB, da Silva JP, E Silva PMR, Martins MA, Linden R. The cellular prion protein modulates phagocytosis and inflammatory response. J Leukoc Biol 2004; 77:238-46. [PMID: 15539455 DOI: 10.1189/jlb.1103531] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The cellular prion protein (PrPc) is a glycoprotein anchored by glycosylphosphatidylinositol (GPI) to the cell surface and is abundantly expressed in the central nervous system. It is also expressed in a variety of cell types of the immune system. We investigated the role of PrPc in the phagocytosis of apoptotic cells and other particles. Macrophages from mice with deletion of the Prnp gene showed higher rates of phagocytosis than wild-type macrophages in in vitro assays. The elimination of GPI-anchored proteins from the cell surface of macrophages from wild-type mice rendered these cells as efficient as macrophages derived from knockout mice. In situ detection of phagocytosis of apoptotic bodies within the retina indicated augmented phagocytotic activity in knockout mice. In an in vivo assay of acute peritonitis, knockout mice showed more efficient phagocytosis of zymosan particles than wild-type mice. In addition, leukocyte recruitment was altered in knockout mice, as compared with wild type. The data show that PrPc modulates phagocytosis in vitro and in vivo. This activity is described for the first time and may be important for normal macrophage functions as well as for the pathogenesis of prion diseases.
Collapse
|
38
|
Rock RB, Gekker G, Hu S, Sheng WS, Cheeran M, Lokensgard JR, Peterson PK. Role of microglia in central nervous system infections. Clin Microbiol Rev 2004; 17:942-64, table of contents. [PMID: 15489356 PMCID: PMC523558 DOI: 10.1128/cmr.17.4.942-964.2004] [Citation(s) in RCA: 512] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The nature of microglia fascinated many prominent researchers in the 19th and early 20th centuries, and in a classic treatise in 1932, Pio del Rio-Hortega formulated a number of concepts regarding the function of these resident macrophages of the brain parenchyma that remain relevant to this day. However, a renaissance of interest in microglia occurred toward the end of the 20th century, fueled by the recognition of their role in neuropathogenesis of infectious agents, such as human immunodeficiency virus type 1, and by what appears to be their participation in other neurodegenerative and neuroinflammatory disorders. During the same period, insights into the physiological and pathological properties of microglia were gained from in vivo and in vitro studies of neurotropic viruses, bacteria, fungi, parasites, and prions, which are reviewed in this article. New concepts that have emerged from these studies include the importance of cytokines and chemokines produced by activated microglia in neurodegenerative and neuroprotective processes and the elegant but astonishingly complex interactions between microglia, astrocytes, lymphocytes, and neurons that underlie these processes. It is proposed that an enhanced understanding of microglia will yield improved therapies of central nervous system infections, since such therapies are, by and large, sorely needed.
Collapse
Affiliation(s)
- R Bryan Rock
- Neuroimmunology Laboratory, Minneapolis Medical Research Foundation, and University of Minnesota Medical School, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Yamaguchi N, Sakaguchi S, Shigematsu K, Okimura N, Katamine S. Doppel-induced Purkinje cell death is stoichiometrically abrogated by prion protein. Biochem Biophys Res Commun 2004; 319:1247-52. [PMID: 15194501 DOI: 10.1016/j.bbrc.2004.05.115] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2004] [Indexed: 11/19/2022]
Abstract
Mice devoid of prion protein (PrP) exhibiting ataxia and Purkinje cell degeneration, such as Ngsk Prnp(-/-) mice, ectopically express PrP-like protein, Dpl, in neurons including Purkinje cells. In this study, two types of transgenic (tg) mice expressing Dpl in neurons, tg(N-Dpl), or Purkinje cells only, tg(P-Dpl), were generated on the background of non-ataxic Zrch I Prnp(-/-) mice. In contrast to the tg mice with the Prnp(+/+) background, both tg mice with the Prnp(-/-) alleles developed Purkinje cell degeneration after incubation periods inversely correlated to the levels of Dpl. Some tg mice hemizygous for Prnp allele also developed disease but much later than those carrying the Prnp(-/-) alleles. This indicates that Dpl expressed by Purkinje cells itself is toxic to the cells, and that the neurotoxicity is stoichiometrically antagonized by PrP.
Collapse
Affiliation(s)
- Naohiro Yamaguchi
- Department of Molecular Microbiology and Immunology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | | | | | | | | |
Collapse
|
40
|
Abstract
The prion protein is a copper (Cu)-binding protein. The abnormal isoform of this protein is associated with the transmissible spongiform encephalopathies or prion diseases. In prion diseases, the prion protein loses its Cu binding capacity. The effect of prion protein expression on the Cu content of the brain was investigated. Transgenic mice, either overexpressing the prion protein or expressing a mutant form lacking the Cu-binding region of the protein, were compared with wild-type mice and mice in which expression of the protein was knocked out. Age-dependent differences in Cu content of the brain were detected. Also, synaptosomal fractions from the brains of the mice showed different Cu content depending on the expression of the prion protein. Mice expressing prion protein, but without the Cu-binding domain showed reduced Cu content. Mice overexpressing the prion protein showed little difference in Cu in the brain compared with wild type but also the prion protein expressed by the mice showed a reduction in the level of Cu bound. These results confirm that prion protein expression modulates the Cu level found at the synapse and this effect is dependent on its Cu binding capacity. Loss of normal Cu binding by the prion protein altered age-related increases in metals in the brain. This may explain why many forms of human prion disease do not develop until late in life.
Collapse
Affiliation(s)
- David R Brown
- Department of Biology and Biochemistry, University of Bath, Cambridge, UK.
| |
Collapse
|
41
|
Abstract
A misfolded version of the prion protein PrP(C), known as PrP(Sc), is the major component of scrapie infectivity, the pathological agent in transmissible spongiform encephalopathies. The Prnp gene that encodes the cellular PrP(C) protein was cloned almost 20 years ago, but remained without sequence or structural relatives for over a decade. Only recently a novel protein, named Doppel (Dpl), was identified, which shares significant biochemical and structural homology with PrP(C). When overexpressed, Dpl is neurotoxic and causes a neurological disease. Strikingly, Dpl neurotoxicity is counteracted and prevented by PrP(C). In contrast to its homologue PrP(C), Dpl is dispensable for prion disease progression and for the generation of PrP(Sc), but Dpl appears to have an essential function in male spermatogenesis. Although Dpl research is still in its infancy, the discovery of Dpl has already solved some enigmas of prion biology and an understanding of its physiological function is emerging.
Collapse
Affiliation(s)
- Axel Behrens
- Mammalian Genetics Laboratory, Cancer Research UK, London, UK
| |
Collapse
|
42
|
Esiri MM, Carter J, Ironside JW. Macrophage and microglial cell immunoreactivity for PrP(c) in a random post-mortem population. Neuropathol Appl Neurobiol 2002; 28:79-82. [PMID: 11849567 DOI: 10.1046/j.1365-2990.2002.00363.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
43
|
Brown DR, Nicholas RSJ, Canevari L. Lack of prion protein expression results in a neuronal phenotype sensitive to stress. J Neurosci Res 2002; 67:211-24. [PMID: 11782965 DOI: 10.1002/jnr.10118] [Citation(s) in RCA: 177] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The prion protein is a highly conserved glycoprotein expressed most highly in the synapse. Evidence has recently been put forward to suggest that the prion protein is an antioxidant. However, the functional importance of the prion protein has been disputed; it is claimed that mice genetically ablated to lack prion protein expression are normal and have no specific phenotype. We have reexamined the phenotype of prion protein knockout mice and found that there are multiple biochemical changes in the mice, including increased levels of nuclear factor NF-kappaB and Mn superoxide dismutase, COX-IV decreased levels of Cu/Zn superoxide dismutase activity, decreased p53, and altered melatonin levels. Additionally, cultured cells from these mice are more sensitive to a range of insults, all linked to increased neuronal sensitivity to oxidative stress. These results imply that prion protein knockout mice are more sensitive to oxidative stress and have an altered phenotype that must be taken into account when considering the additional effects of increased levels of proteins such as Doppel. The implication of these results is that the consequence of genetic ablation of genes must include biochemical analysis as well as analyses of possible developmental and behavioral changes.
Collapse
Affiliation(s)
- David R Brown
- Department of Biochemistry, Cambridge University, Cambridge, United Kingdom.
| | | | | |
Collapse
|
44
|
Abstract
The normal cellular prion protein (PrP(c)) is a membrane sialoglycoprotein of unknown function having the unique property of adopting an abnormal tertiary conformation. The pathological conformer PrP(sc) would be the agent of transmissible spongiform encephalopathies or prion diseases. They include scrapie and bovine spongiform encephalopathy in animals and Creutzfeldt-Jakob disease in humans. The conversion of PrP(c) into PrP(sc) in the brain governs the clinical phenotype of the disease. However, the three-dimensional structure change of PrP(c) can also take place outside the central nervous system, in nonneuronal cells particularly of lymphoid tissue where the agent replicates. In natural infection, PrP(c) in nonneuronal cells of peripheral extracerebral organs may play a key role as the receptor required to enable the entry of the infectious agent into the host. In the present review we have undertaken a first evaluation of compelling data concerning the PrP(c)-expressing cells of nonneuronal origin present in cerebral and extracerebral tissues. The analysis of tissue, cellular, and subcellular localization of PrP(c) may help us better understand the biological function of PrP(c) and provide some information on physiopathological processes underlying prion diseases.
Collapse
Affiliation(s)
- J G Fournier
- Service de Neurovirologie, CEA-DSV/DRM, Fontenay aux Roses, France
| |
Collapse
|
45
|
Burthem J, Urban B, Pain A, Roberts DJ. The normal cellular prion protein is strongly expressed by myeloid dendritic cells. Blood 2001; 98:3733-8. [PMID: 11739179 DOI: 10.1182/blood.v98.13.3733] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Abnormal isoforms of the prion protein (PrP(Sc)) that cause prion diseases are propagated and spread within the body by "carrier" cell(s). Cells of the immune system have been strongly implicated in this process. In particular, PrP(Sc) is known to accumulate on follicular dendritic cells (FDCs) in individuals affected by variant Creutzfeld-Jakob disease. However, FDCs do not migrate widely and the natural history of prion disorders suggests other cells may be required for the transport of PrP(Sc) from the site of ingestion to lymphoid organs and the central nervous system. Substantial evidence suggests that the spread of PrP(Sc) requires bone marrow-derived cells that express normal cellular prion protein (PrP(C)). This study examined the expression of PrP(C) on bone marrow-derived cells that interact with lymphoid follicles. High levels of PrP(C) are present on myeloid dendritic cells (DCs) that surround the splenic white pulp. These myeloid DCs are ontologically and functionally distinct from the FDCs. Consistent with these observations, expression of PrP(C) was strongly induced during the generation of mature myeloid DCs in vitro. In these cells PrP(C) colocalized with major histocompatibility complex class II molecules at the level of light microscopy. Furthermore, given the close anatomic and functional connection of myeloid DCs with lymphoid follicles, these results raise the possibility that myeloid DCs may play a role in the propagation of PrP(Sc) in humans.
Collapse
Affiliation(s)
- J Burthem
- Nuffield Department of Biochemistry and Cellular Science, University of Oxford, Oxford, United Kingdom
| | | | | | | |
Collapse
|
46
|
Daniels M, Cereghetti GM, Brown DR. Toxicity of novel C-terminal prion protein fragments and peptides harbouring disease-related C-terminal mutations. EUROPEAN JOURNAL OF BIOCHEMISTRY 2001; 268:6155-64. [PMID: 11733010 DOI: 10.1046/j.0014-2956.2001.02567.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mice expressing a C-terminal fragment of the prion protein instead of wild-type prion protein die from massive neuronal degeneration within weeks of birth. The C-terminal region of PrPc (PrP121-231) expressed in these mice has an intrinsic neurotoxicity to cultured neurones. Unlike PrPSc, which is not neurotoxic to neurones lacking PrPc expression, PrP121-231 was more neurotoxic to PrPc-deficient cells. Human mutations E200K and F198S were found to enhance toxicity of PrP121-231 to PrP-knockout neurones and E200K enhanced toxicity to wild-type neurones. The normal metabolic cleavage point of PrPc is approximately amino-acid residue 113. A fragment of PrPc corresponding to the whole C-terminus of PrPc (PrP113-231), which is eight amino acids longer than PrP121-231, lacked any toxicity. This suggests the first eight amino residues of PrP113-121 suppress toxicity of the toxic domain in PrP121-231. Addition to cultures of a peptide (PrP112-125) corresponding to this region, in parallel with PrP121-231, suppressed the toxicity of PrP121-231. These results suggest that the prion protein contains two domains that are toxic on their own but which neutralize each other's toxicity in the intact protein. Point mutations in the inherited forms of disease might have their effects by diminishing this inhibition.
Collapse
Affiliation(s)
- M Daniels
- Department of Biochemistry, Cambridge University, UK
| | | | | |
Collapse
|
47
|
Heppner FL, Prinz M, Aguzzi A. Pathogenesis of prion diseases: possible implications of microglial cells. PROGRESS IN BRAIN RESEARCH 2001; 132:737-50. [PMID: 11545032 DOI: 10.1016/s0079-6123(01)32114-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- F L Heppner
- Institute of Neuropathology, Department of Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | | | | |
Collapse
|
48
|
Abstract
Gliosis is one of the hallmarks of the prion diseases. Prion diseases are fatal neurodegenerative conditions of low incidence made famous by both the hypothesis that a protein acts as the infectious agent without involvement of nucleic acid and the speculative idea that a disease of cattle, BSE, has spread to humans from the ingestion of prion-infected beef. Despite these unproved hypotheses, the aetiology of the prion diseases remains unsolved. The rapid degenerative course of the disease is preceded by a long incubation period with little or no symptoms. The rapid neurodegeneration in the disease follows from increased deposition of an abnormal isoform of a normal neuronal protein. Co-incident with the appearance of this abnormal protein is the activation of large numbers of microglia. Studies in cell culture with both the abnormal prion protein and a peptide-mimic suggest that neuronal degeneration occurs because of two concurrent effects. First, there is a reduction in neuronal resistance to toxic insults and, second, there is an increase in the production of toxic substances such as reactive oxygen species by microglia and a decrease in glutamate clearance by astrocytes. Microglia activated by the abnormal form of the prion protein also release cytokines, which stimulate changes in astrocytes such as proliferation. The implication of this is that microglia may play a major role in initiating the pathological changes in prion disease. This review discusses the role of microglia in these changes.
Collapse
Affiliation(s)
- D R Brown
- Department of Biochemistry, Cambridge University, Cambridge, CB2 1QW, United Kingdom.
| |
Collapse
|
49
|
Campbell A, Smith MA, Sayre LM, Bondy SC, Perry G. Mechanisms by which metals promote events connected to neurodegenerative diseases. Brain Res Bull 2001; 55:125-32. [PMID: 11470308 DOI: 10.1016/s0361-9230(01)00455-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although the exact causative phenomenon responsible for the onset and progression of neurodegenerative disorders is at present unresolved, there are some clues as to the mechanisms underlying these chronic diseases. This review addresses mechanisms by which endogenous or environmental factors, through interaction with redox active metals, may initiate a common cascade of events terminating in neurodegeneration.
Collapse
Affiliation(s)
- A Campbell
- Department of Community and Environmental Medicine, University of California, Irvine, Irvine, CA 92697-1825, USA.
| | | | | | | | | |
Collapse
|
50
|
Abstract
The word prion has become synonymous with unusual diseases, such as bovine spongiform encephalopathy and Creutzfeldt-Jakob disease. However, there is also a normal prion protein that does not cause disease. Until recently this highly conserved and widely expressed glycoprotein has been overshadowed by its rogue isoform. Now it is emerging that not only is this protein important for understanding prion disease but it is also important for a healthy brain. The normal cellular isoform of the prion protein is expressed at high levels at synapses suggesting an important role in neuronal function. There is increasing evidence that the normal prion protein binds copper and the resulting complex possesses anti-oxidant activity, and that this, in turn, might have vital implications for synaptic homeostasis.
Collapse
Affiliation(s)
- D R Brown
- Department of Biochemistry, Tennis Court Road, Cambridge University, Cambridge, UK CB2 1QW.
| |
Collapse
|