1
|
Lesion of the olfactory epithelium accelerates prion neuroinvasion and disease onset when prion replication is restricted to neurons. PLoS One 2015; 10:e0119863. [PMID: 25822718 PMCID: PMC4379011 DOI: 10.1371/journal.pone.0119863] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/17/2015] [Indexed: 11/29/2022] Open
Abstract
Natural prion diseases of ruminants are moderately contagious and while the gastrointestinal tract is the primary site of prion agent entry, other mucosae may be entry sites in a subset of infections. In the current study we examined prion neuroinvasion and disease induction following disruption of the olfactory epithelium in the nasal mucosa since this site contains environmentally exposed olfactory sensory neurons that project directly into the central nervous system. Here we provide evidence for accelerated prion neuroinvasion and clinical onset from the olfactory mucosa after disruption and regeneration of the olfactory epithelium and when prion replication is restricted to neurons. In transgenic mice with neuron restricted replication of prions, there was a reduction in survival when the olfactory epithelium was disrupted prior to intranasal inoculation and there was >25% decrease in the prion incubation period. In a second model, the neurotropic DY strain of transmissible mink encephalopathy was not pathogenic in hamsters by the nasal route, but 50% of animals exhibited brain infection and/or disease when the olfactory epithelium was disrupted prior to intranasal inoculation. A time course analysis of prion deposition in the brain following loss of the olfactory epithelium in models of neuron-restricted prion replication suggests that neuroinvasion from the olfactory mucosa is via the olfactory nerve or brain stem associated cranial nerves. We propose that induction of neurogenesis after damage to the olfactory epithelium can lead to prion infection of immature olfactory sensory neurons and accelerate prion spread to the brain.
Collapse
|
2
|
Qualtieri A, Urso E, Pera ML, Sprovieri T, Bossio S, Gambardella A, Quattrone A. Proteomic profiling of cerebrospinal fluid in Creutzfeldt–Jakob disease. Expert Rev Proteomics 2014; 7:907-17. [DOI: 10.1586/epr.10.80] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
3
|
Cho IS, Spinner DS, Kascsak RJ, Meeker HC, Kim BS, Park SY, Schuller-Levis G, Park E. Altered lymphocyte proliferation and innate immune function in scrapie 139A- and ME7-infected mice. Viral Immunol 2013; 26:192-200. [PMID: 23656168 DOI: 10.1089/vim.2012.0091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Lymphoid organs play an important role in prion disease development and progression. While the role of lymphoid organs and changes in immune-related genes have been extensively investigated in scrapie-infected animals, innate immunity has not. Previous studies examined lymphocyte function in scrapie-infected C3H/HeJ mice, which exhibit defects in lipopolysaccharide (LPS) response now known to result from a mutation in Toll-like receptor (TLR) 4. We examined immune function in scrapie-infected CD1 mice, which are LPS responders. Lymphocyte proliferation from CD1 mice infected with either 139A or ME7 scrapie was measured in response to concanavalin (Con) A or LPS at 1 and 3 months after infection. Following LPS exposure, mice infected 3 months with ME7, but not 139A, demonstrated significantly decreased lymphocyte proliferation compared to controls. After Con A exposure, lymphocyte proliferation in scrapie-infected mice did not differ from controls. Gender-specific comparison of lymphocyte proliferation showed significant decreases in mitogenic responses in females infected 3 months with either 139A or ME7, compared to controls. Males infected for 3 months with ME7, but not 139A, showed significantly decreased proliferation after lymphocyte exposure to LPS, but not Con A. Neither gender showed changes in lymphocyte proliferation after 1 month of scrapie infection. Innate immune activation of peritoneal macrophages was determined via production of nitric oxide (NO), IL-6, and TNF-α after exposure to TLR ligands. TNF-α and IL-6 production were reduced in macrophages from females infected with either scrapie strain for 3 months, while NO production after TLR agonist plus IFN-γ exposure was decreased in both females and males infected for 3 months with 139A, compared to ME7. These data demonstrated altered innate immunity, suggesting hormonal and/or other gender-specific regulation may contribute to gender differences in some immune functions. Our data demonstrate lymphocyte proliferation and innate immune functioning in scrapie-infected mice deteriorate with disease progression.
Collapse
Affiliation(s)
- In Soo Cho
- Animal, Plant and Fisheries Quarantine and Inspection Agency, Anyang, Gyunggi-do, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Wisniewski T, Goñi F. Could immunomodulation be used to prevent prion diseases? Expert Rev Anti Infect Ther 2012; 10:307-17. [PMID: 22397565 DOI: 10.1586/eri.11.177] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
All prion diseases are currently without effective treatment and are universally fatal. The underlying pathogenesis of prion diseases (prionoses) is related to an autocatalytic conformational conversion of PrP(C) (C for cellular) to a pathological and infectious conformer known as PrP(Sc) (Sc for scrapie) or PrP(Res) (Res for proteinase K resistant). The past experience with variant Creutzfeldt-Jakob disease, which originated from bovine spongiform encephalopathy, as well as the ongoing epidemic of chronic wasting disease has highlighted the necessity for effective prophylactic and/or therapeutic approaches. Human prionoses are most commonly sporadic, and hence therapy is primarily directed to stop progression; however, in animals the majority of prionoses are infectious and, as a result, the emphasis is on prevention of transmission. These infectious prionoses are most commonly acquired via the alimentary tract as a major portal of infectious agent entry, making mucosal immunization a potentially attractive method to produce a local immune response that can partially or completely prevent prion entry across the gut barrier, while at the same time producing a modulated systemic immunity that is unlikely to be associated with toxicity. A critical factor in any immunomodulatory methodology that targets a self-antigen is the need to delicately balance an effective humoral immune response with potential autoimmune inflammatory toxicity. The ongoing epidemic of chronic wasting disease affecting the USA and Korea, with the potential to spread to human populations, highlights the need for such immunomodulatory approaches.
Collapse
Affiliation(s)
- Thomas Wisniewski
- New York University School of Medicine, 560 First Avenue, New York, NY 10016, USA.
| | | |
Collapse
|
5
|
Enhancement of immunohistochemical staining of scrapie proteins and immune cells within lymph nodes of early scrapie-infected sheep. J Immunol Methods 2011; 371:1-7. [PMID: 21722647 DOI: 10.1016/j.jim.2011.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 06/02/2011] [Accepted: 06/08/2011] [Indexed: 11/23/2022]
Abstract
Transmissible spongiform encephalopathies (TSE) are a group of fatal neurodegenerative diseases that affect animals as well as humans. The oldest of these diseases is Scrapie seen in sheep. Scrapie is caused by an altered form (PrP(sc)), capable of inducing "self-replication" of the normal host prion protein (PrP(c)). There is currently no universal standard for antigen retrieval when using immunohistochemistry to simultaneously stain the PrP(c) protein and other cellular markers. The use of formalin-fixed tissue creates a challenge by concealing the antigenic sites where an antibody would bind, and lengthy antigen retrieval methods must be applied in order to facilitate staining. Further complicating sheep tissue immunohistochemistry is a significant lack of commercial antibodies to sheep cell markers available in research. Here we developed a novel immunohistochemical technique using trypsin, formic acid, and hydrated autoclaving using citraconic anhydride buffer to increase sensitivity of staining for scrapie proteins and immune cell subsets. This allowed us to stain and identify cells within lymphoid tissue associated with early lymphoid pathogenesis in scrapie.
Collapse
|
6
|
Toppets V, Defaweux V, Piret J, Kirschvink N, Grobet L, Antoine N. Features of follicular dendritic cells in ovine pharyngeal tonsil: An in vivo and in vitro study in the context of scrapie pathogenesis. Vet Immunol Immunopathol 2011; 141:26-32. [DOI: 10.1016/j.vetimm.2011.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 01/24/2011] [Accepted: 01/31/2011] [Indexed: 10/18/2022]
|
7
|
Wisniewski T, Goñi F. Immunomodulation for prion and prion-related diseases. Expert Rev Vaccines 2011; 9:1441-52. [PMID: 21105779 DOI: 10.1586/erv.10.131] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Prion diseases are a unique category of illness, affecting both animals and humans, where the underlying pathogenesis is related to a conformational change of a normal self protein called cellular prion protein to a pathological and infectious conformer known as scrapie prion protein (PrP(Sc)). Currently, all prion diseases lack effective treatment and are universally fatal. Past experiences with bovine spongiform encephalopathy and variant Creutzfeldt-Jakob disease mainly in Europe, as well as the current epidemic of chronic wasting disease in North America, have highlighted the need to develop prophylactic and/or therapeutic approaches. In Alzheimer's disease that, like prion disease, is a conformational neurodegenerative disorder, both passive and active immunization has been shown to be highly effective in model animals at preventing disease and cognitive deficits, with emerging data from human trials suggesting that this approach is able to reduce amyloid-related pathology. However, any immunomodulatory approach aimed at a self-antigen has to finely balance an effective humoral immune response with potential autoimmune toxicity. The prion diseases most commonly acquired by infection typically have the alimentary tract as a portal of infectious agent entry. This makes mucosal immunization a potentially attractive method to produce a local immune response that partially or completely prevents prion entry across the gut barrier, while at the same time producing modulated systemic immunity that is unlikely to be associated with toxicity. Our results using an attenuated Salmonella vaccine strain expressing the prion protein showed that mucosal vaccination can protect against prion infection from a peripheral source, suggesting the feasibility of this approach. It is also possible to develop active and/or passive immunomodulatory approaches that more specifically target PrP(Sc) or target the shared pathological conformer found in numerous conformational disorders. Such approaches could have a significant impact on many of the common age-associated dementias.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Department of Psychiatry, Millhauser Laboratories, Room HN419, New York University School of Medicine, 560 First Avenue, New York, NY 10016, USA.
| | | |
Collapse
|
8
|
Fry DE. Prions: reassessment of the germ theory of disease. J Am Coll Surg 2010; 211:546-52. [PMID: 20868977 DOI: 10.1016/j.jamcollsurg.2010.06.389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 06/28/2010] [Accepted: 06/29/2010] [Indexed: 11/28/2022]
Affiliation(s)
- Donald E Fry
- Department of Surgery, Northwestern University Feinberg Schoolof Medicine, Chicago, IL, USA.
| |
Collapse
|
9
|
Kranich J, Krautler NJ, Falsig J, Ballmer B, Li S, Hutter G, Schwarz P, Moos R, Julius C, Miele G, Aguzzi A. Engulfment of cerebral apoptotic bodies controls the course of prion disease in a mouse strain-dependent manner. ACTA ACUST UNITED AC 2010; 207:2271-81. [PMID: 20837697 PMCID: PMC2947076 DOI: 10.1084/jem.20092401] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Progressive accumulation of PrP(Sc), a hallmark of prion diseases, occurs when conversion of PrP(C) into PrP(Sc) is faster than PrP(Sc) clearance. Engulfment of apoptotic bodies by phagocytes is mediated by Mfge8 (milk fat globule epidermal growth factor 8). In this study, we show that brain Mfge8 is primarily produced by astrocytes. Mfge8 ablation induced accelerated prion disease and reduced clearance of cerebellar apoptotic bodies in vivo, as well as excessive PrP(Sc) accumulation and increased prion titers in prion-infected C57BL/6 × 129Sv mice and organotypic cerebellar slices derived therefrom. These phenotypes correlated with the presence of 129Sv genomic markers in hybrid mice and were not observed in inbred C57BL/6 Mfge8(-/-) mice, suggesting the existence of additional strain-specific genetic modifiers. Because Mfge8 receptors are expressed by microglia and depletion of microglia increases PrP(Sc) accumulation in organotypic cerebellar slices, we conclude that engulfment of apoptotic bodies by microglia may be an important pathway of prion clearance controlled by astrocyte-borne Mfge8.
Collapse
Affiliation(s)
- Jan Kranich
- Institute of Neuropathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Transmissible spongiform encephalopathies (TSEs) are inevitably lethal neurodegenerative diseases that affect humans and a large variety of animals. The infectious agent responsible for TSEs is the prion, an abnormally folded and aggregated protein that propagates itself by imposing its conformation onto the cellular prion protein (PrPC) of the host. PrPCis necessary for prion replication and for prion-induced neurodegeneration, yet the proximal causes of neuronal injury and death are still poorly understood. Prion toxicity may arise from the interference with the normal function of PrPC, and therefore, understanding the physiological role of PrPCmay help to clarify the mechanism underlying prion diseases. Here we discuss the evolution of the prion concept and how prion-like mechanisms may apply to other protein aggregation diseases. We describe the clinical and the pathological features of the prion diseases in human and animals, the events occurring during neuroinvasion, and the possible scenarios underlying brain damage. Finally, we discuss potential antiprion therapies and current developments in the realm of prion diagnostics.
Collapse
|
11
|
Nuvolone M, Aguzzi A, Heikenwalder M. Cells and prions: A license to replicate. FEBS Lett 2009; 583:2674-84. [DOI: 10.1016/j.febslet.2009.06.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 06/01/2009] [Accepted: 06/09/2009] [Indexed: 10/20/2022]
|
12
|
Rutishauser D, Mertz KD, Moos R, Brunner E, Rülicke T, Calella AM, Aguzzi A. The comprehensive native interactome of a fully functional tagged prion protein. PLoS One 2009; 4:e4446. [PMID: 19209230 PMCID: PMC2635968 DOI: 10.1371/journal.pone.0004446] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Accepted: 12/15/2008] [Indexed: 11/19/2022] Open
Abstract
The enumeration of the interaction partners of the cellular prion protein, PrP(C), may help clarifying its elusive molecular function. Here we added a carboxy proximal myc epitope tag to PrP(C). When expressed in transgenic mice, PrP(myc) carried a GPI anchor, was targeted to lipid rafts, and was glycosylated similarly to PrP(C). PrP(myc) antagonized the toxicity of truncated PrP, restored prion infectibility of PrP(C)-deficient mice, and was physically incorporated into PrP(Sc) aggregates, indicating that it possessed all functional characteristics of genuine PrP(C). We then immunopurified myc epitope-containing protein complexes from PrP(myc) transgenic mouse brains. Gentle differential elution with epitope-mimetic decapeptides, or a scrambled version thereof, yielded 96 specifically released proteins. Quantitative mass spectrometry with isotope-coded tags identified seven proteins which co-eluted equimolarly with PrP(C) and may represent component of a multiprotein complex. Selected PrP(C) interactors were validated using independent methods. Several of these proteins appear to exert functions in axomyelinic maintenance.
Collapse
Affiliation(s)
- Dorothea Rutishauser
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
- Functional Genomics Center Zurich, Zurich, Switzerland
| | - Kirsten D. Mertz
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Rita Moos
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Erich Brunner
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
- Center for Model Organism Proteomes, University of Zurich, Zurich, Switzerland
| | - Thomas Rülicke
- Institute of Laboratory Animal Science and Research Center Biomodels Austria, University of Veterinary Medicine, Vienna, Austria
| | - Anna Maria Calella
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Hermes G, Ajioka JW, Kelly KA, Mui E, Roberts F, Kasza K, Mayr T, Kirisits MJ, Wollmann R, Ferguson DJP, Roberts CW, Hwang JH, Trendler T, Kennan RP, Suzuki Y, Reardon C, Hickey WF, Chen L, McLeod R. Neurological and behavioral abnormalities, ventricular dilatation, altered cellular functions, inflammation, and neuronal injury in brains of mice due to common, persistent, parasitic infection. J Neuroinflammation 2008; 5:48. [PMID: 18947414 PMCID: PMC2588578 DOI: 10.1186/1742-2094-5-48] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Accepted: 10/23/2008] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Worldwide, approximately two billion people are chronically infected with Toxoplasma gondii with largely unknown consequences. METHODS To better understand long-term effects and pathogenesis of this common, persistent brain infection, mice were infected at a time in human years equivalent to early to mid adulthood and studied 5-12 months later. Appearance, behavior, neurologic function and brain MRIs were studied. Additional analyses of pathogenesis included: correlation of brain weight and neurologic findings; histopathology focusing on brain regions; full genome microarrays; immunohistochemistry characterizing inflammatory cells; determination of presence of tachyzoites and bradyzoites; electron microscopy; and study of markers of inflammation in serum. Histopathology in genetically resistant mice and cytokine and NRAMP knockout mice, effects of inoculation of isolated parasites, and treatment with sulfadiazine or alphaPD1 ligand were studied. RESULTS Twelve months after infection, a time equivalent to middle to early elderly ages, mice had behavioral and neurological deficits, and brain MRIs showed mild to moderate ventricular dilatation. Lower brain weight correlated with greater magnitude of neurologic abnormalities and inflammation. Full genome microarrays of brains reflected inflammation causing neuronal damage (Gfap), effects on host cell protein processing (ubiquitin ligase), synapse remodeling (Complement 1q), and also increased expression of PD-1L (a ligand that allows persistent LCMV brain infection) and CD 36 (a fatty acid translocase and oxidized LDL receptor that mediates innate immune response to beta amyloid which is associated with pro-inflammation in Alzheimer's disease). Immunostaining detected no inflammation around intra-neuronal cysts, practically no free tachyzoites, and only rare bradyzoites. Nonetheless, there were perivascular, leptomeningeal inflammatory cells, particularly contiguous to the aqueduct of Sylvius and hippocampus, CD4+ and CD8+ T cells, and activated microglia in perivascular areas and brain parenchyma. Genetically resistant, chronically infected mice had substantially less inflammation. CONCLUSION In outbred mice, chronic, adult acquired T. gondii infection causes neurologic and behavioral abnormalities secondary to inflammation and loss of brain parenchyma. Perivascular inflammation is prominent particularly contiguous to the aqueduct of Sylvius and hippocampus. Even resistant mice have perivascular inflammation. This mouse model of chronic T. gondii infection raises questions of whether persistence of this parasite in brain can cause inflammation or neurodegeneration in genetically susceptible hosts.
Collapse
Affiliation(s)
- Gretchen Hermes
- Department of Ophthalmology, University of Chicago, Chicago, IL, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Kim HO, Snyder GP, Blazey TM, Race RE, Chesebro B, Skinner PJ. Prion disease induced alterations in gene expression in spleen and brain prior to clinical symptoms. Adv Appl Bioinform Chem 2008; 1:29-50. [PMID: 21918605 PMCID: PMC3169940 DOI: 10.2147/aabc.s3411] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Prion diseases are fatal neurodegenerative disorders that affect animals and humans. There is a need to gain understanding of prion disease pathogenesis and to develop diagnostic assays to detect prion diseases prior to the onset of clinical symptoms. The goal of this study was to identify genes that show altered expression early in the disease process in the spleen and brain of prion disease-infected mice. Using Affymetrix microarrays, we identified 67 genes that showed increased expression in the brains of prion disease-infected mice prior to the onset of clinical symptoms. These genes function in many cellular processes including immunity, the endosome/lysosome system, hormone activity, and the cytoskeleton. We confirmed a subset of these gene expression alterations using other methods and determined the time course in which these changes occur. We also identified 14 genes showing altered expression prior to the onset of clinical symptoms in spleens of prion disease infected mice. Interestingly, four genes, Atp1b1, Gh, Anp32a, and Grn, were altered at the very early time of 46 days post-infection. These gene expression alterations provide insights into the molecular mechanisms underlying prion disease pathogenesis and may serve as surrogate markers for the early detection and diagnosis of prion disease.
Collapse
Affiliation(s)
- Hyeon O Kim
- Department of Veterinary and Biomedical Sciences, University of Minnesota, USA
| | | | | | | | | | | |
Collapse
|
15
|
Aguzzi A, Sigurdson C, Heikenwaelder M. Molecular mechanisms of prion pathogenesis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2008; 3:11-40. [PMID: 18233951 DOI: 10.1146/annurev.pathmechdis.3.121806.154326] [Citation(s) in RCA: 253] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Prion diseases are infectious neurodegenerative diseases occurring in humans and animals with an invariably lethal outcome. One fundamental mechanistic event in prion diseases is the aggregation of aberrantly folded prion protein into large amyloid plaques and fibrous structures associated with neurodegeneration. The cellular prion protein (PrPC) is absolutely required for disease development, and prion knockout mice are not susceptible to prion disease. Prions accumulate not only in the central nervous system but also in lymphoid organs, as shown for new variant and sporadic Creutzfeldt-Jakob patients and for some animals. To date it is largely accepted that prions consist primarily of PrPSc, a misfolded and aggregated beta-sheet-rich isoform of PrPC. However, PrPSc may or may not be completely congruent with the infectious moiety. Here, we discuss the molecular mechanisms leading to neurodegeneration, the role of the immune system in prion pathogenesis, and the existence of prion strains that appear to have different tropisms and biochemical characteristics.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zürich, CH-8091 Zürich, Switzerland.
| | | | | |
Collapse
|
16
|
Abstract
Acquired forms of prion diseases or transmissible spongiform encephalopathies are believed to occur following peripheral exposure. Prions initially accumulate in the lymphoid system before spreading to the nervous system, but the underlying mechanisms for prion transfer between the two systems are still elusive. Here we show that ablation of the B-cell-specific transmembrane protein CD19, a coreceptor of the complement system, results in an acceleration of prion neuroinvasion. This appears to be due to an alteration of the follicular dendritic cell (FDC) network within the lymphoid tissue, thereby reducing the distance between FDCs and adjacent nerve fibers that mediate prion neuroinvasion.
Collapse
|
17
|
Thellung S, Villa V, Corsaro A, Pellistri F, Venezia V, Russo C, Aceto A, Robello M, Florio T. ERK1/2 and p38 MAP kinases control prion protein fragment 90-231-induced astrocyte proliferation and microglia activation. Glia 2007; 55:1469-85. [PMID: 17705195 DOI: 10.1002/glia.20559] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Astrogliosis and microglial activation are a common feature during prion diseases, causing the release of chemoattractant and proinflammatory factors as well as reactive free radicals, involved in neuronal degeneration. The recombinant protease-resistant domain of the prion protein (PrP90-231) displays in vitro neurotoxic properties when refolded in a beta-sheet-rich conformer. Here, we report that PrP90-231 induces the secretion of several cytokines, chemokines, and nitric oxide (NO) release, in both type I astrocytes and microglial cells. PrP90-231 elicited in both cell types the activation of ERK1/2 MAP kinase that displays, in astrocytes, a rapid kinetics and a proliferative response. Conversely, in microglia, PrP90-231-dependent MAP kinase activation was delayed and long lasting, inducing functional activation and growth arrest. In microglial cells, NO release, dependent on the expression of the inducible NO synthase (iNOS), and the secretion of the chemokine CCL5 were Ca(2+) dependent and under the control of the MAP kinases ERK1/2 and p38: ERK1/2 inhibition, using PD98059, reduced iNOS expression, while p38 blockade by PD169316 inhibited CCL5 release. In summary, we demonstrate that glial cells are activated by extracellular misfolded PrP90-231 resulting in a proliferative/secretive response of astrocytes and functional activation of microglia, both dependent on MAP kinase activation. In particular, in microglia, PrP90-231 activated a complex signalling cascade involved in the regulation of NO and chemokine release. These data argue in favor of a causal role for misfolded prion protein in sustaining glial activation and, possibly, glia-mediated neuronal death.
Collapse
Affiliation(s)
- Stefano Thellung
- Department of Oncology, Biology and Genetics, University of Genova, Genova, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Segundo FDS, Sevilla N, Gutiérrez JP, Brun A. Altered lymphocyte homeostasis after oral prion infection in mouse. Vet Immunol Immunopathol 2007; 122:204-15. [PMID: 18207573 DOI: 10.1016/j.vetimm.2007.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Revised: 11/26/2007] [Accepted: 11/30/2007] [Indexed: 01/26/2023]
Abstract
Transmissible spongiform encephalopathies (TSEs) or prion diseases develop as central nervous system (CNS) disorders characterized by extremely long incubation periods. Although TSEs do not go along with inflammatory infiltrates and/or antibody production against the prion protein (PrP(Sc)), the immune system plays an important role in pathogenesis as long as different lymphoid organs (Peyer's patches, lymph nodes and spleen) may facilitate the accumulation and further spread of prions after peripheral exposure. In this work we investigated the changes in lymphoid and dendritic cell (DC) populations as well as the implications of different cytokines during disease progression after experimental oral inoculation of prions in a transgenic mouse model. At different days post-inoculation (dpi), T and B lymphocytes and DC populations from lymphoid organs, blood and brain were analyzed by flow cytometry and immunohistochemistry. Besides time related variations in lymphoid cell numbers due to the aging of the animals significant changes related with the infection were found in mesenteric lymph nodes, peripheral blood leukocytes (PBLs) as well as in spleen, affecting the CD4/CD8 ratio. In contrast, little or no variation was detected in Peyer's Patches or in thymus either associated with aging or the infection status. At individual time points significant differences between infected and control mice were seen in the CD8, CD4 and DC populations, with less evidence of differences in the B cell compartment. Finally, a pro-inflammatory phenotype occurred at early times in the spleen, where the levels of lymphotoxin-beta mRNA were found augmented with respect to controls. Altogether, these results suggest that normal regulation of lymphocyte populations becomes altered along the progression of a prion infection.
Collapse
Affiliation(s)
- F Díaz-San Segundo
- Centro de Investigación en Sanidad Animal (CISA-INIA), Carretera de Algete a El Casar km 8,100, Valdeolmos 28130, Madrid, Spain
| | | | | | | |
Collapse
|
19
|
Heikenwalder M, Federau C, Boehmer LV, Schwarz P, Wagner M, Zeller N, Haybaeck J, Prinz M, Becher B, Aguzzi A. Germinal center B cells are dispensable in prion transport and neuroinvasion. J Neuroimmunol 2007; 192:113-23. [PMID: 17964667 DOI: 10.1016/j.jneuroim.2007.09.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Revised: 09/19/2007] [Accepted: 09/19/2007] [Indexed: 01/17/2023]
Abstract
Transmissible spongiform encephalopathies (TSEs) are fatal neurodegenerative diseases of animals and humans. Many TSEs are initiated by prion replication in the lymphoreticular system (LRS). The cellular and molecular prerequisites for prion trafficking within the LRS are not fully understood. Here we have manipulated CD40 and its ligand to investigate whether genetic or pharmacological ablation of germinal center B cells (GCBs), which migrate into and out of germinal centers, influences prion pathogenesis. In contrast to previous reports, no alteration of prion pathogenesis was detected in mice lacking CD40L and in mice treated with anti-CD40L antibodies. These results suggest that GCBs alone do not impact peripheral splenic prion transport, replication efficiency, or neuroinvasion, and point to other mechanisms affecting prion transport from lymphoreticular sites of replication to the nervous system.
Collapse
Affiliation(s)
- Mathias Heikenwalder
- Institute of Neuropathology, University Hospital of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Dorban G, Defaweux V, Demonceau C, Flandroy S, Van Lerberghe PB, Falisse-Poirrier N, Piret J, Heinen E, Antoine N. Interaction between dendritic cells and nerve fibres in lymphoid organs after oral scrapie exposure. Virchows Arch 2007; 451:1057-65. [PMID: 17823814 DOI: 10.1007/s00428-007-0476-6] [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] [Received: 05/09/2007] [Revised: 07/12/2007] [Accepted: 07/13/2007] [Indexed: 01/16/2023]
Abstract
In transmissible spongiform encephalopathies (TSEs), the infectious agent, called PrPsc, an abnormal isoform of the cellular prion protein, accumulates and replicates in lymphoid organs before affecting the nervous system. To clarify the cellular requirements for the neuroinvasion of the scrapie agent from the lymphoid organs to the central nervous system, we have studied, by confocal microscopy, the innervations within Peyer's patches, mesenteric lymph nodes and the spleen of mice in physiological conditions and after oral exposure to prion. Contacts between nerve fibres and PrPsc-associated cells, dendritic cells (DCs) and follicular dendritic cells (FDCs), were evaluated in preclinical prion-infected mice. Using a double immunolabelling strategy, we demonstrated the lack of innervation of PrPsc-accumulating cells (FDCs). Contacts between nerve fibers and PrPsc-propagating cells (DCs) were detected in T-cell zones and cell-trafficking areas. This supports, for the first time, the possible implication of dendritic cells in the prion neuroinvasion process.
Collapse
Affiliation(s)
- Gauthier Dorban
- Human Histology, Immunology Center, Faculty of Medicine, University of Liège, CHU, Avenue de l'hôpital, Tour de pharmacie +4, 4000 Liège, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Prion Diseases. Neurobiol Dis 2007. [DOI: 10.1016/b978-012088592-3/50044-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
22
|
Abstract
The infectious particle causing transmissible spongiform encephalopathy (TSE), a fatal neurodegenerative disease of humans and animals, has been termed prion. Its major component is an aggregated variant of the cellular prion protein, PrP(C). The main target of prion pathology is the central nervous system (CNS), yet most prion diseases are initiated or accompanied by prion replication at extracerebral locations, including secondary lymphoid organs, muscle and, in some instances, blood. How do prions travel from the periphery into the CNS? Is this an active or a passive process and does neuronal prion transport explain the long incubation times in prion diseases? Alternatively, if prion infectivity arises spontaneously in the CNS, as believed from sporadic Creutzfeldt-Jakob patients, how do prions manage to travel from the CNS into the periphery (e.g., spleen, muscle) of the infected host? The mechanisms of neuronal prion transport from the periphery into the CNS or vice versa are heavily investigated and debated but poorly understood. Although research in the past has accumulated knowledge on prion progression from the periphery to the brain, we are far from understanding the molecular mechanisms of neuronal prion transport.
Collapse
Affiliation(s)
- Mathias Heikenwalder
- Department of Pathology, Institute of Neuropathology, University Hospital of Zürich, Zürich, Switzerland.
| | | | | |
Collapse
|
23
|
Priller J, Prinz M, Heikenwalder M, Zeller N, Schwarz P, Heppner FL, Aguzzi A. Early and rapid engraftment of bone marrow-derived microglia in scrapie. J Neurosci 2006; 26:11753-62. [PMID: 17093096 PMCID: PMC6674798 DOI: 10.1523/jneurosci.2275-06.2006] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Prion neuroinvasion is accompanied by maximal activation of microglia, the significance of which for pathogenesis is unknown. Here, we used bone marrow (BM) cells expressing GFP (green fluorescent protein) to study the turnover of microglia in mouse scrapie. We found that >or=50% of all brain microglia were replaced by BM-derived cells before clinical disease onset. In terminally sick mice, microglia density increased threefold to fourfold. Hence BM-derived microglia rapidly and efficaciously colonize the brain in scrapie. Whereas reconstitution of wild-type mice with prion protein-deficient (Prnp(o/o)) BM did not alter scrapie pathogenesis, Prnp(o/o) mice transplanted with wild-type BM cells were resistant to peripherally administered prions despite high levels of infectivity in the spleen. Cerebellar homogenates from prion-inoculated Prnp(o/o) mice reconstituted with >10% of wild-type microglia failed to infect transgenic mice overexpressing the cellular prion protein. Hence, in contrast to previous reports, microglia are not competent for efficient prion transport and replication in vivo.
Collapse
Affiliation(s)
- Josef Priller
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
- Departments of Psychiatry and Experimental Neurology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany, and
| | - Marco Prinz
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
- Institute of Neuropathology, University of Göttingen, 37075 Göttingen, Germany
| | - Mathias Heikenwalder
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
| | - Nicolas Zeller
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
| | - Petra Schwarz
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
| | - Frank L. Heppner
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
24
|
Aguzzi A, Heikenwalder M. Pathogenesis of prion diseases: current status and future outlook. Nat Rev Microbiol 2006; 4:765-75. [PMID: 16980938 DOI: 10.1038/nrmicro1492] [Citation(s) in RCA: 164] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The prion, a conformational variant of a host protein, is the infectious particle responsible for transmissible spongiform encephalopathy (TSE), a fatal neurodegenerative disease of humans and animals. The principal target of prion pathology is the brain, yet most TSEs also display prion replication at extra-cerebral locations, including secondary lymphoid organs and sites of chronic inflammation. Despite significant progress in our understanding of this infectious agent, many fundamental questions relating to the nature of the prion, including the mechanism of replication and the molecular events underlying brain damage, remain unanswered. Here we focus on the unresolved issues pertaining to prion pathogenesis, particularly on the role played by the immune system.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zürich, Schmelzbergstrasse 12, CH-8091 Zürich, Switzerland.
| | | |
Collapse
|
25
|
Davies GA, Bryant AR, Reynolds JD, Jirik FR, Sharkey KA. Prion diseases and the gastrointestinal tract. CANADIAN JOURNAL OF GASTROENTEROLOGY = JOURNAL CANADIEN DE GASTROENTEROLOGIE 2006; 20:18-24. [PMID: 16432555 PMCID: PMC2538961 DOI: 10.1155/2006/184528] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The gastrointestinal (GI) tract plays a central role in the pathogenesis of transmissible spongiform encephalopathies. These are human and animal diseases that include bovine spongiform encephalopathy, scrapie and Creutzfeldt-Jakob disease. They are uniformly fatal neurological diseases, which are characterized by ataxia and vacuolation in the central nervous system. Although they are known to be caused by the conversion of normal cellular prion protein to its infectious conformational isoform (PrPsc) the process by which this isoform is propagated and transported to the brain remains poorly understood. M cells, dendritic cells and possibly enteroendocrine cells are important in the movement of infectious prions across the GI epithelium. From there, PrPsc propagation requires B lymphocytes, dendritic cells and follicular dendritic cells of Peyer's patches. The early accumulation of the disease-causing agent in the plexuses of the enteric nervous system supports the contention that the autonomic nervous system is important in disease transmission. This is further supported by the presence of PrPsc in the ganglia of the parasympathetic and sympathetic nerves that innervate the GI tract. Additionally, the lymphoreticular system has been implicated as the route of transmission from the gut to the brain. Although normal cellular prion protein is found in the enteric nervous system, its role has not been characterized. Further research is required to understand how the cellular components of the gut wall interact to propagate and transmit infectious prions to develop potential therapies that may prevent the progression of transmissible spongiform encephalopathies.
Collapse
Affiliation(s)
- Gwynivere A Davies
- Institute for Infection, Immunity and Inflammation, University of Calgary, Calgary, Alberta
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta
- Department of Physiology and Biophysics, University of Calgary, Calgary, Alberta
| | - Adam R Bryant
- Institute for Infection, Immunity and Inflammation, University of Calgary, Calgary, Alberta
- Department of Anatomy and Cell Biology, University of Calgary, Calgary, Alberta
| | - John D Reynolds
- Institute for Infection, Immunity and Inflammation, University of Calgary, Calgary, Alberta
- Department of Anatomy and Cell Biology, University of Calgary, Calgary, Alberta
| | - Frank R Jirik
- Alberta Bone and Joint Institute, University of Calgary, Calgary, Alberta
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta
| | - Keith A Sharkey
- Institute for Infection, Immunity and Inflammation, University of Calgary, Calgary, Alberta
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta
- Department of Physiology and Biophysics, University of Calgary, Calgary, Alberta
- Correspondence: Dr Keith Sharkey, Department of Physiology and Biophysics, University of Calgary, 3330 Hospital Drive Northwest, Calgary, Alberta T2N 4N1. Telephone 403–220–4601, fax 403–283–3028, e-mail
| |
Collapse
|
26
|
Drayton DL, Liao S, Mounzer RH, Ruddle NH. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol 2006; 7:344-53. [PMID: 16550197 DOI: 10.1038/ni1330] [Citation(s) in RCA: 527] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The development of lymphoid organs can be viewed as a continuum. At one end are the 'canonical' secondary lymphoid organs, including lymph nodes and spleen; at the other end are 'ectopic' or tertiary lymphoid organs, which are cellular accumulations arising during chronic inflammation by the process of lymphoid neogenesis. Secondary lymphoid organs are genetically 'preprogrammed' and 'prepatterned' during ontogeny, whereas tertiary lymphoid organs arise under environmental influences and are not restricted to specific developmental 'windows' or anatomic locations. Between these two boundaries are other types of lymphoid tissues that are less developmentally but more environmentally regulated, such as Peyer's patches, nasal-associated lymphoid tissue, bronchial-associated lymphoid tissue and inducible bronchial-associated lymphoid tissue. Their regulation, functions and potential effects are discussed here.
Collapse
Affiliation(s)
- Danielle L Drayton
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520-8034, USA
| | | | | | | |
Collapse
|
27
|
Bergström AL, Jensen TK, Heegaard PMH, Cordes H, Hansen VB, Laursen H, Lind P. Short-term Study of the Uptake of PrPSc by the Peyer's Patches in Hamsters after Oral Exposure to Scrapie. J Comp Pathol 2006; 134:126-33. [PMID: 16473365 DOI: 10.1016/j.jcpa.2005.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2005] [Accepted: 08/11/2005] [Indexed: 10/25/2022]
Abstract
The disease-associated prion protein (PrP(Sc)) has been detected in the ileal Peyer's patches of lambs as early as one week after oral exposure to scrapie. In hamsters, the earliest reported time of PrP(Sc) detection in the Peyer's patches after oral exposure to scrapie is 69 days post-infection. To evaluate the acute uptake of inoculum and to investigate whether the Peyer's patches constitute the primary site of entry for scrapie after oral exposure, hamsters were each exposed orally to 1 ml of a 10% brain homogenate from hamsters in the terminal stage of infection with the 263 K strain of the scrapie agent. PrP(Sc) was demonstrated in the Peyer's patches only a few days after exposure, i.e., much earlier than previously reported. This study supports the view that the Peyer's patches constitute at least one of the primary entry sites of PrP(Sc) after oral exposure to scrapie.
Collapse
Affiliation(s)
- A-L Bergström
- Department of Veterinary Diagnostics, The Danish Institute for Food and Veterinary Research, Bülowsvej 27, 1790 Copenhagen V, Denmark
| | | | | | | | | | | | | |
Collapse
|
28
|
Seeger H, Heikenwalder M, Zeller N, Kranich J, Schwarz P, Gaspert A, Seifert B, Miele G, Aguzzi A. Coincident Scrapie Infection and Nephritis Lead to Urinary Prion Excretion. Science 2005; 310:324-6. [PMID: 16224026 DOI: 10.1126/science.1118829] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Prion infectivity is typically restricted to the central nervous and lymphatic systems of infected hosts, but chronic inflammation can expand the distribution of prions. We tested whether chronic inflammatory kidney disorders would trigger excretion of prion infectivity into urine. Urinary proteins from scrapie-infected mice with lymphocytic nephritis induced scrapie upon inoculation into noninfected indicator mice. Prionuria was found in presymptomatic scrapie-infected and in sick mice, whereas neither prionuria nor urinary PrP(Sc) was detectable in prion-infected wild-type or PrP(C)-overexpressing mice, or in nephritic mice inoculated with noninfectious brain. Thus, urine may provide a vector for horizontal prion transmission, and inflammation of excretory organs may influence prion spread.
Collapse
Affiliation(s)
- Harald Seeger
- Institute of Neuropathology, University Hospital of Zürich, Schmelzbergstrasse 12, CH-8091 Zürich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|