1
|
Pal R, Thomas CM, Salamat K, Jenkins SJ, Bradford BM, Mabbott NA. Acute LPS exposure enhances susceptibility to peripheral prion infection. Sci Rep 2025; 15:9754. [PMID: 40119036 PMCID: PMC11928655 DOI: 10.1038/s41598-025-94003-3] [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: 12/03/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
After peripheral infections, the initial accumulation of prions within secondary lymphoid tissues is essential for the transmission of disease to the brain. Macrophages are considered to sequester or destroy prions, but little was known of their impact on disease susceptibility after a peripheral infection. Inflammation in the peritoneal cavity can trigger the macrophage disappearance reaction, whereby the macrophages are temporarily contained within cellular aggregates on the mesothelium. We studied the impact of the bacterial lipopolysaccharide (LPS)-mediated macrophage disappearance reaction on susceptibility to an intraperitoneal prion infection. Intraperitoneal LPS injection significantly enhanced prion disease susceptibility approximately 100X when given 24-3 h before infection. The effects on disease susceptibility coincided with the reduced abundance of macrophages within the peritoneal cavity at the time of infection and the enhanced early accumulation of prions in the spleen. This suggests that the reduced recoverable abundance of macrophages in the peritoneal cavity following acute LPS-treatment, increased disease susceptibility by enhancing the initial propagation of the prions from site of exposure (peritoneal cavity) to the spleen from where they subsequently spread to the brain. Further studies may help identify novel macrophage-targeted treatments that can reduce susceptibility to peripherally acquired prion infections.
Collapse
Affiliation(s)
- Reiss Pal
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Charlotte M Thomas
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Khalid Salamat
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Stephen J Jenkins
- Queens Medical Research Institute, University of Edinburgh Centre for Inflammation Research, Edinburgh, UK
| | - Barry M Bradford
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK.
| | - Neil A Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK.
| |
Collapse
|
2
|
Chang SC, Arifin MI, Tahir W, McDonald KJ, Zeng D, Schatzl HM, Hannaoui S, Gilch S. Extraneural infection route restricts prion conformational variability and attenuates the impact of quaternary structure on infectivity. PLoS Pathog 2024; 20:e1012370. [PMID: 38976748 PMCID: PMC11257401 DOI: 10.1371/journal.ppat.1012370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/18/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024] Open
Abstract
Prions can exist as different strains that consist of conformational variants of the misfolded, pathogenic prion protein isoform PrPSc. Defined by stably transmissible biological and biochemical properties, strains have been identified in a spectrum of prion diseases, including chronic wasting disease (CWD) of wild and farmed cervids. CWD is highly contagious and spreads via direct and indirect transmission involving extraneural sites of infection, peripheral replication and neuroinvasion of prions. Here, we investigated the impact of infection route on CWD prion conformational selection and propagation. We used gene-targeted mouse models expressing deer PrP for intracerebral or intraperitoneal inoculation with fractionated or unfractionated brain homogenates from white-tailed deer, harboring CWD strains Wisc-1 or 116AG. Upon intracerebral inoculation, Wisc-1 and 116AG-inoculated mice differed in conformational stability of PrPSc. In brains of mice infected intraperitoneally with either inoculum, PrPSc propagated with identical conformational stability and fewer PrPSc deposits in most brain regions than intracerebrally inoculated animals. For either inoculum, PrPSc conformational stability in brain and spinal cord was similar upon intracerebral infection but significantly higher in spinal cords of intraperitoneally infected animals. Inoculation with fractionated brain homogenates resulted in lower variance of survival times upon intraperitoneal compared to intracerebral infection. In summary, we demonstrate that extraneural infection mitigates the impact of PrPSc quaternary structure on infection and reduces conformational variability of PrPSc propagated in the brain. These findings provide new insights into the evolution of stable CWD strains in natural, extraneural transmissions.
Collapse
Affiliation(s)
- Sheng Chun Chang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | | | - Waqas Tahir
- Canadian and WOAH Reference Laboratory for BSE, Canadian Food Inspection Agency, Lethbridge, Canada
| | | | - Doris Zeng
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Hermann M. Schatzl
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| | - Samia Hannaoui
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Sabine Gilch
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
3
|
Miyahara H, Dai J, Li Y, Cui X, Takeuchi H, Hachiya N, Kametani F, Yazaki M, Mori M, Higuchi K. Macrophages in the reticuloendothelial system inhibit early induction stages of mouse apolipoprotein A-II amyloidosis. Amyloid 2022:1-14. [PMID: 36495239 DOI: 10.1080/13506129.2022.2153667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Amyloidosis refers to a group of degenerative diseases that are characterized by the deposition of misfolded protein fibrils in various organs. Deposited amyloid may be removed by a phagocyte-dependent innate immune system; however, the precise mechanisms during disease progression remain unclear. We herein investigated the properties of macrophages that contribute to amyloid degradation and disease progression using inducible apolipoprotein A-II amyloidosis model mice. Intravenously injected AApoAII amyloid was efficiently engulfed by reticuloendothelial macrophages in the liver and spleen and disappeared by 24 h. While cultured murine macrophages degraded AApoAII via the endosomal-lysosomal pathway, AApoAII fibrils reduced cell viability and phagocytic capacity. Furthermore, the depletion of reticuloendothelial macrophages before the induction of AApoAII markedly increased hepatic and splenic AApoAII deposition. These results highlight the physiological role of reticuloendothelial macrophages in the early stages of pathogenesis and suggest the maintenance of phagocytic integrity as a therapeutic strategy to inhibit disease progression.
Collapse
Affiliation(s)
- Hiroki Miyahara
- Department of Neuro-Health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Jian Dai
- Department of Neuro-Health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Ying Li
- Department of Aging Biology, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Xiaoran Cui
- Department of Aging Biology, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Hibiki Takeuchi
- Department of Aging Biology, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | | | - Fuyuki Kametani
- Department of Brain and Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masahide Yazaki
- Department of Neuro-Health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Masayuki Mori
- Department of Neuro-Health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan.,Department of Aging Biology, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Keiichi Higuchi
- Department of Neuro-Health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan.,Department of Aging Biology, Shinshu University Graduate School of Medicine, Matsumoto, Japan.,Community Health Care Research Center, Nagano University of Health and Medicine, Nagano, Japan
| |
Collapse
|
4
|
Bradford BM, McGuire LI, Hume DA, Pridans C, Mabbott NA. Microglia deficiency accelerates prion disease but does not enhance prion accumulation in the brain. Glia 2022; 70:2169-2187. [PMID: 35852018 PMCID: PMC9544114 DOI: 10.1002/glia.24244] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 01/08/2023]
Abstract
Prion diseases are transmissible, neurodegenerative disorders associated with misfolding of the prion protein. Previous studies show that reduction of microglia accelerates central nervous system (CNS) prion disease and increases the accumulation of prions in the brain, suggesting that microglia provide neuroprotection by phagocytosing and destroying prions. In Csf1rΔFIRE mice, the deletion of an enhancer within Csf1r specifically blocks microglia development, however, their brains develop normally and show none of the deficits reported in other microglia-deficient models. Csf1rΔFIRE mice were used as a refined model in which to study the impact of microglia-deficiency on CNS prion disease. Although Csf1rΔFIRE mice succumbed to CNS prion disease much earlier than wild-type mice, the accumulation of prions in their brains was reduced. Instead, astrocytes displayed earlier, non-polarized reactive activation with enhanced phagocytosis of neuronal contents and unfolded protein responses. Our data suggest that rather than simply phagocytosing and destroying prions, the microglia instead provide host-protection during CNS prion disease and restrict the harmful activities of reactive astrocytes.
Collapse
Affiliation(s)
- Barry M. Bradford
- The Roslin Institute and R(D)SVSUniversity of Edinburgh, Easter Bush CampusMidlothianUK
| | - Lynne I. McGuire
- The Roslin Institute and R(D)SVSUniversity of Edinburgh, Easter Bush CampusMidlothianUK
| | - David A. Hume
- Mater Research Institute‐University of Queensland, Translational Research InstituteWoolloongabbaQueenslandAustralia
| | - Clare Pridans
- Simons Initiative for the Developing Brain, Centre for Discovery Brain SciencesUniversity of Edinburgh, Hugh Robson BuildingEdinburghUK
- Centre for Inflammation ResearchThe Queen's Medical Research Institute, Edinburgh BioQuarterEdinburghUK
| | - Neil A. Mabbott
- The Roslin Institute and R(D)SVSUniversity of Edinburgh, Easter Bush CampusMidlothianUK
| |
Collapse
|
5
|
Mammadova N, Cassmann ED, Moore SJ, Nicholson EM, Greenlee JJ. Experimental inoculation of CD11c + B1 lymphocytes, CD68 + macrophages, or platelet-rich plasma from scrapie-infected sheep into susceptible sheep results in variable infectivity. Access Microbiol 2020; 2:acmi000155. [PMID: 33195984 PMCID: PMC7656192 DOI: 10.1099/acmi.0.000155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/24/2020] [Indexed: 11/18/2022] Open
Abstract
Many studies have demonstrated prion infectivity in whole blood and blood components in a variety of transmissible spongiform encephalopathies of livestock and rodents, and variant Creutzfeldt–Jakob disease in humans, as well as an association between pathogenic prion protein (PrPSc) and different immune cells (e.g. follicular dendritic cells, T and B lymphocytes, monocytes and tingible body macrophages). To further investigate the role of various blood components in prion disease transmission, we intracranially inoculated genetically susceptible VRQ/ARQ and ARQ/ARQ sheep with inocula composed of CD11c+ B1 lymphocytes, CD68 +macrophages, or platelet-rich plasma derived from clinically ill sheep infected with the US no. 13–7 scrapie agent. At the completion of the study, we found that VRQ/ARQ and ARQ/ARQ sheep inoculated with CD11c+ B1 lymphocytes and CD68+ macrophages developed scrapie with detectable levels of PrPSc in the central nervous system and lymphoreticular system, while those inoculated with platelet-rich plasma did not develop disease and did not have detectable PrPSc by immunohistochemistry or enzyme immunoassay. This study complements and expands on earlier findings that white blood cells harbour prion infectivity, and reports CD11c+ B1 lymphocytes and CD68+ macrophages as additional targets for possible preclinical detection of prion infection in blood.
Collapse
Affiliation(s)
- Najiba Mammadova
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA.,Oak Ridge Institute for Science and Education (ORISE) through an interagency agreement between the U.S. Department of Energy (DOE) and the U.S. Department of Agriculture (USDA). ORISE is managed by ORAU under DOE contract number DE-SC0014664
| | - Eric D Cassmann
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA.,Oak Ridge Institute for Science and Education (ORISE) through an interagency agreement between the U.S. Department of Energy (DOE) and the U.S. Department of Agriculture (USDA). ORISE is managed by ORAU under DOE contract number DE-SC0014664
| | - S Jo Moore
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA.,Oak Ridge Institute for Science and Education (ORISE) through an interagency agreement between the U.S. Department of Energy (DOE) and the U.S. Department of Agriculture (USDA). ORISE is managed by ORAU under DOE contract number DE-SC0014664
| | - Eric M Nicholson
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Justin J Greenlee
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| |
Collapse
|
6
|
Mabbott NA, Bradford BM, Pal R, Young R, Donaldson DS. The Effects of Immune System Modulation on Prion Disease Susceptibility and Pathogenesis. Int J Mol Sci 2020; 21:E7299. [PMID: 33023255 PMCID: PMC7582561 DOI: 10.3390/ijms21197299] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Prion diseases are a unique group of infectious chronic neurodegenerative disorders to which there are no cures. Although prion infections do not stimulate adaptive immune responses in infected individuals, the actions of certain immune cell populations can have a significant impact on disease pathogenesis. After infection, the targeting of peripherally-acquired prions to specific immune cells in the secondary lymphoid organs (SLO), such as the lymph nodes and spleen, is essential for the efficient transmission of disease to the brain. Once the prions reach the brain, interactions with other immune cell populations can provide either host protection or accelerate the neurodegeneration. In this review, we provide a detailed account of how factors such as inflammation, ageing and pathogen co-infection can affect prion disease pathogenesis and susceptibility. For example, we discuss how changes to the abundance, function and activation status of specific immune cell populations can affect the transmission of prion diseases by peripheral routes. We also describe how the effects of systemic inflammation on certain glial cell subsets in the brains of infected individuals can accelerate the neurodegeneration. A detailed understanding of the factors that affect prion disease transmission and pathogenesis is essential for the development of novel intervention strategies.
Collapse
Affiliation(s)
- Neil A. Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (B.M.B.); (R.P.); (R.Y.); (D.S.D.)
| | | | | | | | | |
Collapse
|
7
|
Effect of co-infection with a small intestine-restricted helminth pathogen on oral prion disease pathogenesis in mice. Sci Rep 2019; 9:6674. [PMID: 31040320 PMCID: PMC6491469 DOI: 10.1038/s41598-019-42900-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/11/2019] [Indexed: 11/24/2022] Open
Abstract
The early replication of some orally-acquired prion strains upon stromal-derived follicular dendritic cells (FDC) within the small intestinal Peyer’s patches is essential to establish host infection, and for the disease to efficiently spread to the brain. Factors that influence the early accumulation of prions in Peyer’s patches can directly influence disease pathogenesis. The host’s immune response to a gastrointestinal helminth infection can alter susceptibility to co-infection with certain pathogenic bacteria and viruses. Here we used the natural mouse small intestine-restricted helminth pathogen Heligmosomoides polygyrus to test the hypothesis that pathology specifically within the small intestine caused by a helminth co-infection would influence oral prion disease pathogenesis. When mice were co-infected with prions on d 8 after H. polygyrus infection the early accumulation of prions within Peyer’s patches was reduced and survival times significantly extended. Natural prion susceptible hosts such as sheep, deer and cattle are regularly exposed to gastrointestinal helminth parasites. Our data suggest that co-infections with small intestine-restricted helminth pathogens may be important factors that influence oral prion disease pathogenesis.
Collapse
|
8
|
|
9
|
Mabbott NA. How do PrP Sc Prions Spread between Host Species, and within Hosts? Pathogens 2017; 6:pathogens6040060. [PMID: 29186791 PMCID: PMC5750584 DOI: 10.3390/pathogens6040060] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/16/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022] Open
Abstract
Prion diseases are sub-acute neurodegenerative diseases that affect humans and some domestic and free-ranging animals. Infectious prion agents are considered to comprise solely of abnormally folded isoforms of the cellular prion protein known as PrPSc. Pathology during prion disease is restricted to the central nervous system where it causes extensive neurodegeneration and ultimately leads to the death of the host. The first half of this review provides a thorough account of our understanding of the various ways in which PrPSc prions may spread between individuals within a population, both horizontally and vertically. Many natural prion diseases are acquired peripherally, such as by oral exposure, lesions to skin or mucous membranes, and possibly also via the nasal cavity. Following peripheral exposure, some prions accumulate to high levels within the secondary lymphoid organs as they make their journey from the site of infection to the brain, a process termed neuroinvasion. The replication of PrPSc prions within secondary lymphoid organs is important for their efficient spread to the brain. The second half of this review describes the key tissues, cells and molecules which are involved in the propagation of PrPSc prions from peripheral sites of exposure (such as the lumen of the intestine) to the brain. This section also considers how additional factors such as inflammation and aging might influence prion disease susceptibility.
Collapse
Affiliation(s)
- Neil A Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| |
Collapse
|
10
|
Mabbott NA. Immunology of Prion Protein and Prions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:203-240. [PMID: 28838662 DOI: 10.1016/bs.pmbts.2017.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Many natural prion diseases are acquired peripherally, such as following the oral consumption of contaminated food or pasture. After peripheral exposure many prion isolates initially accumulate to high levels within the host's secondary lymphoid tissues. The replication of prions within these tissues is essential for their efficient spread to the brain where they ultimately cause neurodegeneration. This chapter describes our current understanding of the critical tissues, cells, and molecules which the prions exploit to mediate their efficient propagation from the site of exposure (such as the intestine) to the brain. Interactions between the immune system and prions are not only restricted to the secondary lymphoid tissues. Therefore, an account of how the activation status of the microglial in the brain can also influence progression of prion disease pathogenesis is provided. Prion disease susceptibility may also be influenced by additional factors such as chronic inflammation, coinfection with other pathogens, and aging. Finally, the potential for immunotherapy to provide a means of safe and effective prophylactic or therapeutic intervention in these currently untreatable diseases is considered.
Collapse
Affiliation(s)
- Neil A Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Midlothian, United Kingdom.
| |
Collapse
|
11
|
Oral Prion Disease Pathogenesis Is Impeded in the Specific Absence of CXCR5-Expressing Dendritic Cells. J Virol 2017; 91:JVI.00124-17. [PMID: 28275192 PMCID: PMC5411578 DOI: 10.1128/jvi.00124-17] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/02/2017] [Indexed: 01/09/2023] Open
Abstract
After oral exposure, the early replication of certain prion strains upon stromal cell-derived follicular dendritic cells (FDC) in the Peyer's patches in the small intestine is essential for the efficient spread of disease to the brain. However, little is known of how prions are initially conveyed from the gut lumen to establish infection on FDC. Our previous data suggest that mononuclear phagocytes such as CD11c+ conventional dendritic cells play an important role in the initial propagation of prions from the gut lumen into Peyer's patches. However, whether these cells conveyed orally acquired prions toward FDC within Peyer's patches was not known. The chemokine CXCL13 is expressed by FDC and follicular stromal cells and modulates the homing of CXCR5-expressing cells toward the FDC-containing B cell follicles. Here, novel compound transgenic mice were created in which a CXCR5 deficiency was specifically restricted to CD11c+ cells. These mice were used to determine whether CXCR5-expressing conventional dendritic cells propagate prions toward FDC after oral exposure. Our data show that in the specific absence of CXCR5-expressing conventional dendritic cells the early accumulation of prions upon FDC in Peyer's patches and the spleen was impaired, and disease susceptibility significantly reduced. These data suggest that CXCR5-expressing conventional dendritic cells play an important role in the efficient propagation of orally administered prions toward FDC within Peyer's patches in order to establish host infection.IMPORTANCE Many natural prion diseases are acquired by oral consumption of contaminated food or pasture. Once the prions reach the brain they cause extensive neurodegeneration, which ultimately leads to death. In order for the prions to efficiently spread from the gut to the brain, they first replicate upon follicular dendritic cells within intestinal Peyer's patches. How the prions are first delivered to follicular dendritic cells to establish infection was unknown. Understanding this process is important since treatments which prevent prions from infecting follicular dendritic cells can block their spread to the brain. We created mice in which mobile conventional dendritic cells were unable to migrate toward follicular dendritic cells. In these mice the early accumulation of prions on follicular dendritic cells was impaired and oral prion disease susceptibility was reduced. This suggests that prions exploit conventional dendritic cells to facilitate their initial delivery toward follicular dendritic cells to establish host infection.
Collapse
|
12
|
Donaldson DS, Sehgal A, Rios D, Williams IR, Mabbott NA. Increased Abundance of M Cells in the Gut Epithelium Dramatically Enhances Oral Prion Disease Susceptibility. PLoS Pathog 2016; 12:e1006075. [PMID: 27973593 PMCID: PMC5156364 DOI: 10.1371/journal.ppat.1006075] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/17/2016] [Indexed: 02/07/2023] Open
Abstract
Many natural prion diseases of humans and animals are considered to be acquired through oral consumption of contaminated food or pasture. Determining the route by which prions establish host infection will identify the important factors that influence oral prion disease susceptibility and to which intervention strategies can be developed. After exposure, the early accumulation and replication of prions within small intestinal Peyer's patches is essential for the efficient spread of disease to the brain. To replicate within Peyer's patches, the prions must first cross the gut epithelium. M cells are specialised epithelial cells within the epithelia covering Peyer's patches that transcytose particulate antigens and microorganisms. M cell-development is dependent upon RANKL-RANK-signalling, and mice in which RANK is deleted only in the gut epithelium completely lack M cells. In the specific absence of M cells in these mice, the accumulation of prions within Peyer's patches and the spread of disease to the brain was blocked, demonstrating a critical role for M cells in the initial transfer of prions across the gut epithelium in order to establish host infection. Since pathogens, inflammatory stimuli and aging can modify M cell-density in the gut, these factors may also influence oral prion disease susceptibility. Mice were therefore treated with RANKL to enhance M cell density in the gut. We show that prion uptake from the gut lumen was enhanced in RANKL-treated mice, resulting in shortened survival times and increased disease susceptibility, equivalent to a 10-fold higher infectious titre of prions. Together these data demonstrate that M cells are the critical gatekeepers of oral prion infection, whose density in the gut epithelium directly limits or enhances disease susceptibility. Our data suggest that factors which alter M cell-density in the gut epithelium may be important risk factors which influence host susceptibility to orally acquired prion diseases.
Collapse
Affiliation(s)
- David S. Donaldson
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, United Kingdom
| | - Anuj Sehgal
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, United Kingdom
| | - Daniel Rios
- Dept. Pathology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Ifor R. Williams
- Dept. Pathology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Neil A. Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Donaldson DS, Mabbott NA. The influence of the commensal and pathogenic gut microbiota on prion disease pathogenesis. J Gen Virol 2016; 97:1725-1738. [PMID: 27193137 DOI: 10.1099/jgv.0.000507] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Prion diseases are a unique group of transmissible, chronic, neurodegenerative disorders. Following peripheral exposure (e.g. oral), prions often accumulate first within the secondary lymphoid tissues before they infect the central nervous system (CNS). Prion replication within secondary lymphoid tissues is crucial for the efficient spread of disease to the CNS. Once within the CNS, the responses of innate immune cells within it can have a significant influence on neurodegeneration and disease progression. Recently, there have been substantial advances in our understanding of how cross-talk between the host and the vast community of commensal microorganisms present at barrier surfaces such as the gut influences the development and regulation of the host's immune system. These effects are evident not only in the mucosal immune system in the gut, but also in the CNS. The actions of this microbial community (the microbiota) have many important beneficial effects on host health, from metabolism of nutrients and regulation of host development to protection from pathogen infection. However, the microbiota can also have detrimental effects in some circumstances. In this review we discuss the many and varied interactions between prions, the host and the gut microbiota. Particular emphasis is given to the ways by which changes to the composition of the commensal gut microbiota or congruent pathogen infection may influence prion disease pathogenesis and/or disease susceptibility. Understanding how these factors influence prion pathogenesis and disease susceptibility is important for assessing the risk to infection and the design of novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- David S Donaldson
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, UK
| | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
14
|
The Good, the Bad, and the Ugly of Dendritic Cells during Prion Disease. J Immunol Res 2015; 2015:168574. [PMID: 26697507 PMCID: PMC4677227 DOI: 10.1155/2015/168574] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 11/15/2015] [Indexed: 12/11/2022] Open
Abstract
Prions are a unique group of proteinaceous pathogens which cause neurodegenerative disease and can be transmitted by a variety of exposure routes. After peripheral exposure, the accumulation and replication of prions within secondary lymphoid organs are obligatory for their efficient spread from the periphery to the brain where they ultimately cause neurodegeneration and death. Mononuclear phagocytes (MNP) are a heterogeneous population of dendritic cells (DC) and macrophages. These cells are abundant throughout the body and display a diverse range of roles based on their anatomical locations. For example, some MNP are strategically situated to provide a first line of defence against pathogens by phagocytosing and destroying them. Conventional DC are potent antigen presenting cells and migrate via the lymphatics to the draining lymphoid tissue where they present the antigens to lymphocytes. The diverse roles of MNP are also reflected in various ways in which they interact with prions and in doing so impact on disease pathogenesis. Indeed, some studies suggest that prions exploit conventional DC to infect the host. Here we review our current understanding of the influence of MNP in the pathogenesis of the acquired prion diseases with particular emphasis on the role of conventional DC.
Collapse
|
15
|
Bellingham SA, Guo B, Hill AF. The secret life of extracellular vesicles in metal homeostasis and neurodegeneration. Biol Cell 2015; 107:389-418. [PMID: 26032945 DOI: 10.1111/boc.201500030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/27/2015] [Indexed: 12/21/2022]
Abstract
Biologically active metals such as copper, zinc and iron are fundamental for sustaining life in different organisms with the regulation of cellular metal homeostasis tightly controlled through proteins that coordinate metal uptake, efflux and detoxification. Many of the proteins involved in either uptake or efflux of metals are localised and function on the plasma membrane, traffic between intracellular compartments depending upon the cellular metal environment and can undergo recycling via the endosomal pathway. The biogenesis of exosomes also occurs within the endosomal system, with several major neurodegenerative disease proteins shown to be released in association with these vesicles, including the amyloid-β (Aβ) peptide in Alzheimer's disease and the infectious prion protein involved in Prion diseases. Aβ peptide and the prion protein also bind biologically active metals and are postulated to play important roles in metal homeostasis. In this review, we will discuss the role of extracellular vesicles in Alzheimer's and Prion diseases and explore their potential contribution to metal homeostasis.
Collapse
Affiliation(s)
- Shayne A Bellingham
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Belinda Guo
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Andrew F Hill
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Extracellular vesicles--Their role in the packaging and spread of misfolded proteins associated with neurodegenerative diseases. Semin Cell Dev Biol 2015; 40:89-96. [PMID: 25704308 DOI: 10.1016/j.semcdb.2015.02.007] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 02/13/2015] [Accepted: 02/15/2015] [Indexed: 12/13/2022]
Abstract
Many cell types, including neurons, are known to release small membranous vesicles known as exosomes. In addition to their protein content these vesicles have recently been shown to contain messenger RNA (mRNA) and micro RNA (miRNA) species. Roles for these vesicles include cell-cell signalling, removal of unwanted proteins, and transfer of pathogens (including prion-like misfolded proteins) between cells, such as infectious prions. Prions are the infectious particles that are responsible for transmissible neurodegenerative diseases such as Creutzfeldt-Jakob disease (CJD) of humans or bovine spongiform encephalopathy (BSE) of cattle. Exosomes are also involved in processing the amyloid precursor protein (APP), which is associated with Alzheimer's disease (AD). As exosomes can be isolated from circulating fluids such as serum, urine, and cerebrospinal fluid (CSF), they provide a potential source of biomarkers for neurological conditions. Here, we review the roles these vesicles play in neurodegenerative disease and highlight their potential in diagnosing these disorders through analysis of their RNA content.
Collapse
|
17
|
Sakudo A, Onodera T. Prion protein (PrP) gene-knockout cell lines: insight into functions of the PrP. Front Cell Dev Biol 2015; 2:75. [PMID: 25642423 PMCID: PMC4295555 DOI: 10.3389/fcell.2014.00075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/22/2014] [Indexed: 11/13/2022] Open
Abstract
Elucidation of prion protein (PrP) functions is crucial to fully understand prion diseases. A major approach to studying PrP functions is the use of PrP gene-knockout (Prnp (-/-)) mice. So far, six types of Prnp (-/-) mice have been generated, demonstrating the promiscuous functions of PrP. Recently, other PrP family members, such as Doppel and Shadoo, have been found. However, information obtained from comparative studies of structural and functional analyses of these PrP family proteins do not fully reveal PrP functions. Recently, varieties of Prnp (-/-) cell lines established from Prnp (-/-) mice have contributed to the analysis of PrP functions. In this mini-review, we focus on Prnp (-/-) cell lines and summarize currently available Prnp (-/-) cell lines and their characterizations. In addition, we introduce the recent advances in the methodology of cell line generation with knockout or knockdown of the PrP gene. We also discuss how these cell lines have provided valuable insights into PrP functions and show future perspectives.
Collapse
Affiliation(s)
- Akikazu Sakudo
- Laboratory of Biometabolic Chemistry, Faculty of Medicine, School of Health Sciences, University of the Ryukyus Nishihara, Japan
| | - Takashi Onodera
- Research Center for Food Safety, School of Agricultural and Life Sciences, University of Tokyo Tokyo, Japan
| |
Collapse
|
18
|
Klöhn PC, Castro-Seoane R, Collinge J. Exosome release from infected dendritic cells: a clue for a fast spread of prions in the periphery? J Infect 2013; 67:359-68. [PMID: 23911964 DOI: 10.1016/j.jinf.2013.07.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/11/2013] [Accepted: 07/23/2013] [Indexed: 11/18/2022]
Abstract
Prion diseases are incurable transmissible neurological disorders. In many natural and experimental prion diseases, infectious prions can be detected in the lymphoreticular system (LRS) long before they reach the brain where they cause a fatal rapidly progressive degeneration. Although major cell types that contribute to prion accumulation have been identified, the mode of prion dissemination in the LRS remains elusive. Recent evidence of a remarkably fast splenic prion accumulation after peripheral infection of mice, resulting in high prion titers in dendritic cells (DCs) and a release of prions from infected DCs via exosomes suggest that intercellular dissemination may contribute to rapid prion colonization in the LRS. A vast body of evidence from retroviral infections shows that DCs and other antigen-presenting cells (APCs) share viral antigens by intercellular transfer to warrant immunity against viruses if APCs remain uninfected. Evolved to adapt the immune response to evading pathogens, these pathways may constitute a portal for unimpeded prion dissemination owing to the tolerance of the immune system against host-encoded prion protein. In this review we summarize current paradigms for antigen-sharing pathways which may be relevant to better understand dissemination of rogue neurotoxic proteins.
Collapse
Affiliation(s)
- Peter-Christian Klöhn
- MRC Prion Unit and Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| | | | | |
Collapse
|
19
|
Mabbott NA. Prion pathogenesis and secondary lymphoid organs (SLO): tracking the SLO spread of prions to the brain. Prion 2012; 6:322-33. [PMID: 22895090 PMCID: PMC3609058 DOI: 10.4161/pri.20676] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Prion diseases are subacute neurodegenerative diseases that affect humans and a range of domestic and free-ranging animal species. These diseases are characterized by the accumulation of PrPSc, an abnormally folded isoform of the cellular prion protein (PrPC), in affected tissues. The pathology during prion disease appears to occur almost exclusively within the central nervous system. The extensive neurodegeneration which occurs ultimately leads to the death of the host. An intriguing feature of the prion diseases, when compared with other protein-misfolding diseases, is their transmissibility. Following peripheral exposure, some prion diseases accumulate to high levels within lymphoid tissues. The replication of prions within lymphoid tissue has been shown to be important for the efficient spread of disease to the brain. This article describes recent progress in our understanding of the cellular mechanisms that influence the propagation of prions from peripheral sites of exposure (such as the lumen of the intestine) to the brain. A thorough understanding of these events will lead to the identification of important targets for therapeutic intervention, or alternatively, reveal additional processes that influence disease susceptibility to peripherally-acquired prion diseases.
Collapse
Affiliation(s)
- Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, Midlothian, UK.
| |
Collapse
|
20
|
Piercey Åkesson C, Press CM, Tranulis MA, Jeffrey M, Aleksandersen M, Landsverk T, Espenes A. Phenotypic characterization of cells participating in transport of prion protein aggregates across the intestinal mucosa of sheep. Prion 2012; 6:261-75. [PMID: 22437736 PMCID: PMC3399537 DOI: 10.4161/pri.19215] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The oral route is considered to be the main entry site of several transmissible spongiform encephalopathies or prion diseases of animals and man. Following natural and experimental oral exposure to scrapie, sheep first accumulate disease associated prion protein (PrPd) in Peyer’s patch (PP) lymphoid follicles. In this study, recombinant ovine prion protein (rPrP) was inoculated into gut loops of young lambs and the transportation across the intestinal wall studied. In particular, the immunohistochemical phenotypes of cells bearing the inoculated prion protein were investigated. The rPrP was shown to be transported across the villi of the gut, into the lacteals and submucosal lymphatics, mimicking the transport route of PrPd from scrapie brain inoculum observed in a previous intestinal loop experiment. The cells bearing the inoculated rPrP were mainly mononuclear cells, and multicolor immunofluorescence procedures were used to show that the rPrP bearing cells were professional antigen presenting cells expressing Major histocompatibility complex II (MHCII). In addition, the rPrP bearing cells labeled with CD205, CD11b and the macrophage marker CD68, and not with the dendritic cell markers CD11c and CD209. Others have reported that cells expressing CD205 and CD11b in the absence of CD11c have been shown to induce T cell tolerance or regulatory T cells. Based on this association, it was speculated that the rPrP and by extension PrPd and scrapie infective material may exploit the physiological process of macromolecular uptake across the gut, and that this route of entry may have implications for immune surveillance.
Collapse
Affiliation(s)
- Caroline Piercey Åkesson
- Department of Basic Sciences and Aquatic Medicine, Norwegian School of Veterinary Science, Oslo, Norway.
| | | | | | | | | | | | | |
Collapse
|
21
|
Awaad A, Nakamura M, Ishimura K. Histochemical and biochemical analysis of the size-dependent nanoimmunoresponse in mouse Peyer's patches using fluorescent organosilica particles. Int J Nanomedicine 2012; 7:1423-39. [PMID: 22619503 PMCID: PMC3356209 DOI: 10.2147/ijn.s28675] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background/objective The size-dependent mucosal immunoresponse against nanomaterials (nanoimmunoresponse) is an important approach for mucosal vaccination. In the present work, the size-dependent nanoimmunoresponse of mouse Peyer’s patches (PPs) and immunoglobulin A (IgA) level was investigated using fluorescent thiol-organosilica particles. Methods Various sizes of fluorescent thiol-organosilica particles (100, 180, 365, 745, and 925 nm in diameter) were administered orally. PPs were analyzed histochemically, and IgA levels in PP homogenates, intestinal secretions around PPs, and bile were analyzed biochemically. Results When compared with the larger particles (745 and 925 nm), oral administration of smaller thiol-organosilica particles (100, 180, and 365 nm) increased the number of CD11b+ macrophages and IgA+ cells in the subepithelial domes of the PPs. Additionally, administration of larger particles induced the expression of alpha-L-fucose and mucosal IgA on the surface of M cells in the follicle-associated epithelia of PPs and increased the number of 33D1+ dendritic cells in the subepithelial domes of the PPs. IgA contents in the bile and PP homogenates were high after the administration of the 100 nm particles, but IgA levels in the intestinal secretions were high after the administration of the 925 nm particles. Two size-dependent routes of IgA secretions into the intestinal lumen, the enterohepatic route for smaller particles and the mucosal route for larger particles were proposed. Conclusion Thiol-organosilica particles demonstrated size-dependent nanoimmunoresponse after oral administration. The size of the particles may control the mucosal immunity in PPs and were useful in mucosal vaccination approaches.
Collapse
Affiliation(s)
- Aziz Awaad
- Department of Anatomy and Cell Biology, the University of Tokushima Graduate School, Kuramoto, Tokushima, Japan
| | | | | |
Collapse
|
22
|
Castro-Seoane R, Hummerich H, Sweeting T, Tattum MH, Linehan JM, Fernandez de Marco M, Brandner S, Collinge J, Klöhn PC. Plasmacytoid dendritic cells sequester high prion titres at early stages of prion infection. PLoS Pathog 2012; 8:e1002538. [PMID: 22359509 PMCID: PMC3280992 DOI: 10.1371/journal.ppat.1002538] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 01/04/2012] [Indexed: 12/22/2022] Open
Abstract
In most transmissible spongiform encephalopathies prions accumulate in the lymphoreticular system (LRS) long before they are detectable in the central nervous system. While a considerable body of evidence showed that B lymphocytes and follicular dendritic cells play a major role in prion colonization of lymphoid organs, the contribution of various other cell types, including antigen-presenting cells, to the accumulation and the spread of prions in the LRS are not well understood. A comprehensive study to compare prion titers of candidate cell types has not been performed to date, mainly due to limitations in the scope of animal bioassays where prohibitively large numbers of mice would be required to obtain sufficiently accurate data. By taking advantage of quantitative in vitro prion determination and magnetic-activated cell sorting, we studied the kinetics of prion accumulation in various splenic cell types at early stages of prion infection. Robust estimates for infectious titers were obtained by statistical modelling using a generalized linear model. Whilst prions were detectable in B and T lymphocytes and in antigen-presenting cells like dendritic cells and macrophages, highest infectious titers were determined in two cell types that have previously not been associated with prion pathogenesis, plasmacytoid dendritic (pDC) and natural killer (NK) cells. At 30 days after infection, NK cells were more than twice, and pDCs about seven-fold, as infectious as lymphocytes respectively. This result was unexpected since, in accordance to previous reports prion protein, an obligate requirement for prion replication, was undetectable in pDCs. This underscores the importance of prion sequestration and dissemination by antigen-presenting cells which are among the first cells of the immune system to encounter pathogens. We furthermore report the first evidence for a release of prions from lymphocytes and DCs of scrapie-infected mice ex vivo, a process that is associated with a release of exosome-like membrane vesicles.
Collapse
Affiliation(s)
- Rocio Castro-Seoane
- MRC Prion Unit and Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Holger Hummerich
- MRC Prion Unit and Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Trevor Sweeting
- Department of Statistical Science, University College London, London, United Kingdom
| | - M. Howard Tattum
- MRC Prion Unit and Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Jacqueline M. Linehan
- MRC Prion Unit and Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Mar Fernandez de Marco
- MRC Prion Unit and Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Sebastian Brandner
- MRC Prion Unit and Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - John Collinge
- MRC Prion Unit and Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Peter-Christian Klöhn
- MRC Prion Unit and Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| |
Collapse
|
23
|
Natale G, Pasquali L, Paparelli A, Fornai F. Parallel manifestations of neuropathologies in the enteric and central nervous systems. Neurogastroenterol Motil 2011; 23:1056-65. [PMID: 21951862 DOI: 10.1111/j.1365-2982.2011.01794.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Neurodegenerative diseases may extend outside the central nervous system (CNS) and involve the gastrointestinal (GI) tract. The gut would appear to be a pathological marker for neurodegeneration, as well as a site for studying the pathophysiology of neurodegeneration. In fact, both in the ENS and CNS, misfolded proteins are likely to initiate a process of neurodegeneration. For example, the very same protein aggregates can be detected both in the ENS and CNS. In both systems, misfolded proteins are likely to share common cell-to-cell diffusion mechanisms, which may occur through a parallel prion-like diffusion process. Independently from the enteric or central origin, misfolded proteins may proceed along the following steps, they: (i) form aggregates; (ii) are expressed on plasma membrane; (iii) are secreted extracellularly; (iv) are glycated to form advanced glycation end-products (AGEs); (v) are internalized through specific receptors placed on neighboring cells (RAGEs); (vi) are cleared by autophagy; and (vii) are neurotoxic. These features are common for a-synuclein (in Parkinson's disease and other synucleinopathies), β-amyloid and tau (in degenerative dementia), SOD-1 and TDP43 (in amyotrophic lateral sclerosis), and PrPsc (in prion diseases). While in some diseases these features are common to both ENS and CNS, in others this remains a working hypothesis. PURPOSE This review analyzes GI alterations from a pathological perspective to assess whether the enteric nervous system (ENS) mirrors the neuropathology described in the CNS. We discuss the potential mechanisms that lead to the onset and spread of neurodegeneration within the gut, from the gut to the brain, and vice versa.
Collapse
Affiliation(s)
- G Natale
- Department of Human Morphology and Applied Biology, University of Pisa, Italy
| | | | | | | |
Collapse
|
24
|
McCulloch L, Brown KL, Bradford BM, Hopkins J, Bailey M, Rajewsky K, Manson JC, Mabbott NA. Follicular dendritic cell-specific prion protein (PrP) expression alone is sufficient to sustain prion infection in the spleen. PLoS Pathog 2011; 7:e1002402. [PMID: 22144895 PMCID: PMC3228802 DOI: 10.1371/journal.ppat.1002402] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 10/11/2011] [Indexed: 11/20/2022] Open
Abstract
Prion diseases are characterised by the accumulation of PrP(Sc), an abnormally folded isoform of the cellular prion protein (PrP(C)), in affected tissues. Following peripheral exposure high levels of prion-specific PrP(Sc) accumulate first upon follicular dendritic cells (FDC) in lymphoid tissues before spreading to the CNS. Expression of PrP(C) is mandatory for cells to sustain prion infection and FDC appear to express high levels. However, whether FDC actively replicate prions or simply acquire them from other infected cells is uncertain. In the attempts to-date to establish the role of FDC in prion pathogenesis it was not possible to dissociate the Prnp expression of FDC from that of the nervous system and all other non-haematopoietic lineages. This is important as FDC may simply acquire prions after synthesis by other infected cells. To establish the role of FDC in prion pathogenesis transgenic mice were created in which PrP(C) expression was specifically "switched on" or "off" only on FDC. We show that PrP(C)-expression only on FDC is sufficient to sustain prion replication in the spleen. Furthermore, prion replication is blocked in the spleen when PrP(C)-expression is specifically ablated only on FDC. These data definitively demonstrate that FDC are the essential sites of prion replication in lymphoid tissues. The demonstration that Prnp-ablation only on FDC blocked splenic prion accumulation without apparent consequences for FDC status represents a novel opportunity to prevent neuroinvasion by modulation of PrP(C) expression on FDC.
Collapse
Affiliation(s)
- Laura McCulloch
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Midlothian, United Kingdom
| | - Karen L. Brown
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Midlothian, United Kingdom
| | - Barry M. Bradford
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Midlothian, United Kingdom
| | - John Hopkins
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Midlothian, United Kingdom
| | - Mick Bailey
- Division of Veterinary Pathology, Infection and Immunity, School of Clinical Veterinary Science, University of Bristol, Avon, United Kingdom
| | - Klaus Rajewsky
- Program in Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jean C. Manson
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Midlothian, United Kingdom
| | - Neil A. Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
25
|
Natale G, Ferrucci M, Lazzeri G, Paparelli A, Fornai F. Transmission of prions within the gut and towards the central nervous system. Prion 2011; 5:142-9. [PMID: 21814041 DOI: 10.4161/pri.5.3.16328] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The prion protein is a glycoprotein characterized by a folded α-helical structure that, under pathological conditions, misfolds and aggregates into its infectious isoform as β-sheet rich amyloidic deposits. The accumulation of the abnormal protein is responsible for a group of progressive and fatal disorders characterized by vacuolation, gliosis, and spongiform degeneration. Prion disorders are characterized by a triple aetiology: familial, sporadic or acquired, although most cases are sporadic. The mechanisms underlying prion neurotoxicity remain controversial, while novel findings lead to hypothesize intriguing pathways responsible for prion spreading. The present review aims to examine the involvement of the gastrointestinal tract and hypothesizes the potential mechanisms underlying cell-to-cell transmission of the prion protein. In particular, a special emphasis is posed on the mechanisms of prion transmission within the gut and towards the central nervous system. The glycation of prion protein to form advanced glycation end-products (AGE) interacting with specific receptors placed on neighboring cells (RAGE) represents the key hypothesis to be discussed.
Collapse
Affiliation(s)
- Gianfranco Natale
- Department of Human Morphology and Applied Biology, University of Pisa, Pisa, Italy
| | | | | | | | | |
Collapse
|
26
|
Okada H, Iwamaru Y, Imamura M, Masujin K, Matsuura Y, Murayama Y, Mohri S, Yokoyama T. Detection of disease-associated prion protein in the posterior portion of the small intestine involving the continuous Peyer's patch in cattle orally infected with bovine spongiform encephalopathy agent. Transbound Emerg Dis 2011; 58:333-43. [PMID: 21320296 DOI: 10.1111/j.1865-1682.2011.01208.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Twenty-eight calves were exposed to 5 g of homogenized brainstems confirmed as bovine spongiform encephalopathy (BSE) agents. Two to five animals were sequentially killed for post-mortem analyses 20 months post-inoculation (MPI) at intervals of 6 or 12 months. Samples from animals challenged orally with BSE agents were examined by Western blot and immunohistochemical analyses. Immunolabelled, disease-associated prion protein (PrPsc) was detected in a small portion of follicles in the continuous Peyer's patch from the posterior portion of the small intestine involving the entire ileum and the posterior jejunum but not in the discrete Peyer's patches in the remaining jejunum in preclinical animals at 20, 36, and 48 MPI. The PrPsc-positive cells corresponded to tingible body macrophages on double immunofluorescence labelling. In addition, PrPsc accumulated in 7 of 14 animals in the central nervous system (CNS) after 34 MPI, and five of them developed clinical signs and were killed at 34, 46, 58, and 66 MPI. Two preclinical animals killed at 36 and 48 MPI presented the earliest detectable and smallest deposition of immunolabelled PrPsc in the dorsal motor nucleus of the vagus nerve, the spinal trigeminal nucleus of the medulla oblongata at the obex region, and/or the intermediolateral nucleus of the 13th thoracic segment of the spinal cord. Based on serial killing, no PrPsc was detectable in the CNS, including the medulla oblongata at the obex level, before 30 MPI, by Western blot and immunohistochemical analyses. These results are important for understanding the pathogenesis of BSE.
Collapse
Affiliation(s)
- H Okada
- Prion Disease Research Center, National Institute of Animal Health, Tsukuba, Ibaraki, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Almeida LM, Basu U, Khaniya B, Taniguchi M, Williams JL, Moore SS, Guan LL. Gene expression in the medulla following oral infection of cattle with bovine spongiform encephalopathy. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2011; 74:110-126. [PMID: 21218340 DOI: 10.1080/15287394.2011.529061] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The identification of variations in gene expression in response to bovine spongiform encephalopathy (BSE) may help to elucidate the mechanisms of neuropathology and prion replication and discover biomarkers for disease. In this study, genes that are differentially expressed in the caudal medulla tissues of animals infected with different doses of PrP(BSE) at 12 and 45 mo post infection were compared using array containing 24,000 oligonucleotide probes. Data analysis identified 966 differentially expressed (DE) genes between control and infected animals. Genes identified in at least two of four experiments (control versus 1-g infected animals at 12 and 45-mo; control versus 100-g infected animals at 12 and 45 mo) were considered to be the genes that may be associated with BSE disease. From the 176 DE genes associated with BSE, 84 had functions described in the Gene Ontology (GO) database. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of 14 genes revealed that prion infection may cause dysfunction of several different networks, including extracellular matrix (ECM), cell adhesion, neuroactive ligand-receptor interaction, complement and coagulation cascades, MAPK signaling, neurodegenerative disorder, SNARE interactions in vesicular transport, and the transforming growth factor (TGF) beta signaling pathways. The identification of DE genes will contribute to a better understanding of the molecular mechanisms of neuropathology in bovine species. Additional studies on larger number of animals are in progress in our laboratory to investigate the roles of these DE genes in pathogenesis of BSE.
Collapse
Affiliation(s)
- Luciane M Almeida
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
28
|
Transcytosis of murine-adapted bovine spongiform encephalopathy agents in an in vitro bovine M cell model. J Virol 2010; 84:12285-91. [PMID: 20861256 DOI: 10.1128/jvi.00969-10] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Transmissible spongiform encephalopathies (TSE), including bovine spongiform encephalopathy (BSE), are fatal neurodegenerative disorders in humans and animals. BSE appears to have spread to cattle through the consumption of feed contaminated with BSE/scrapie agents. In the case of an oral infection, the agents have to cross the gut-epithelial barrier. We recently established a bovine intestinal epithelial cell line (BIE cells) that can differentiate into the M cell type in vitro after lymphocytic stimulation (K. Miyazawa, T. Hondo, T. Kanaya, S. Tanaka, I. Takakura, W. Itani, M. T. Rose, H. Kitazawa, T. Yamaguchi, and H. Aso, Histochem. Cell Biol. 133:125-134, 2010). In this study, we evaluated the role of M cells in the intestinal invasion of the murine-adapted BSE (mBSE) agent using our in vitro bovine intestinal epithelial model. We demonstrate here that M cell-differentiated BIE cells are able to transport the mBSE agent without inactivation at least 30-fold more efficiently than undifferentiated BIE cells in our in vitro model. As M cells in the follicle-associated epithelium are known to have a high ability to transport a variety of macromolecules, viruses, and bacteria from gut lumen to mucosal immune cells, our results indicate the possibility that bovine M cells are able to deliver agents of TSE, not just the mBSE agent.
Collapse
|
29
|
Mathiason CK, Hayes-Klug J, Hays SA, Powers J, Osborn DA, Dahmes SJ, Miller KV, Warren RJ, Mason GL, Telling GC, Young AJ, Hoover EA. B cells and platelets harbor prion infectivity in the blood of deer infected with chronic wasting disease. J Virol 2010; 84:5097-107. [PMID: 20219916 PMCID: PMC2863796 DOI: 10.1128/jvi.02169-09] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 02/23/2010] [Indexed: 11/20/2022] Open
Abstract
Substantial evidence for prion transmission via blood transfusion exists for many transmissible spongiform encephalopathy (TSE) diseases. Determining which cell phenotype(s) is responsible for trafficking infectivity has important implications for our understanding of the dissemination of prions, as well as their detection and elimination from blood products. We used bioassay studies of native white-tailed deer and transgenic cervidized mice to determine (i) if chronic wasting disease (CWD) blood infectivity is associated with the cellular versus the cell-free/plasma fraction of blood and (ii) in particular if B-cell (MAb 2-104(+)), platelet (CD41/61(+)), or CD14(+) monocyte blood cell phenotypes harbor infectious prions. All four deer transfused with the blood mononuclear cell fraction from CWD(+) donor deer became PrP(CWD) positive by 19 months postinoculation, whereas none of the four deer inoculated with cell-free plasma from the same source developed prion infection. All four of the deer injected with B cells and three of four deer receiving platelets from CWD(+) donor deer became PrP(CWD) positive in as little as 6 months postinoculation, whereas none of the four deer receiving blood CD14(+) monocytes developed evidence of CWD infection (immunohistochemistry and Western blot analysis) after 19 months of observation. Results of the Tg(CerPrP) mouse bioassays mirrored those of the native cervid host. These results indicate that CWD blood infectivity is cell associated and suggest a significant role for B cells and platelets in trafficking CWD infectivity in vivo and support earlier tissue-based studies associating putative follicular B cells with PrP(CWD). Localization of CWD infectivity with leukocyte subpopulations may aid in enhancing the sensitivity of blood-based diagnostic assays for CWD and other TSEs.
Collapse
Affiliation(s)
- Candace K. Mathiason
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - Jeanette Hayes-Klug
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - Sheila A. Hays
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - Jenny Powers
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - David A. Osborn
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - Sallie J. Dahmes
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - Karl V. Miller
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - Robert J. Warren
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - Gary L. Mason
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - Glenn C. Telling
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - Alan J. Young
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| | - Edward A. Hoover
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, National Park Service, Fort Collins, Colorado, Warnell School of Forestry and Natural Resources, University of Georgia, Athens, Georgia, WASCO Inc., Monroe, Georgia, University of Kentucky Medical Center, Lexington, Kentucky, South Dakota State University, Brookings, South Dakota
| |
Collapse
|
30
|
Lwin S, Inoshima Y, Atoji Y, Ueno H, Ishiguro N. Immune cell types involved in early uptake and transport of recombinant mouse prion protein in Peyer’s patches of calves. Cell Tissue Res 2009; 338:343-54. [DOI: 10.1007/s00441-009-0879-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Accepted: 09/07/2009] [Indexed: 12/23/2022]
|
31
|
Brown KL, Wathne GJ, Sales J, Bruce ME, Mabbott NA. The effects of host age on follicular dendritic cell status dramatically impair scrapie agent neuroinvasion in aged mice. THE JOURNAL OF IMMUNOLOGY 2009; 183:5199-207. [PMID: 19786551 DOI: 10.4049/jimmunol.0802695] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Following peripheral exposure, many transmissible spongiform encephalopathy (TSE) agents accumulate first in lymphoid tissues before spreading to the CNS (termed neuroinvasion) where they cause neurodegeneration. Early TSE agent accumulation upon follicular dendritic cells (FDCs) in lymphoid follicles appears critical for efficient neuroinvasion. Most clinical cases of variant Creutzfeldt-Jakob disease have occurred in young adults, although the reasons behind this apparent age-related susceptibility are uncertain. Host age has a significant influence on immune function. As FDC status and immune complex trapping is reduced in aged mice (600 days old), we hypothesized that this aging-related decline in FDC function might impair TSE pathogenesis. We show that coincident with the effects of host age on FDC status, the early TSE agent accumulation in the spleens of aged mice was significantly impaired. Furthermore, following peripheral exposure, none of the aged mice developed clinical TSE disease during their lifespans, although most mice displayed histopathological signs of TSE disease in their brains. Our data imply that the reduced status of FDCs in aged mice significantly impairs the early TSE agent accumulation in lymphoid tissues and subsequent neuroinvasion. Furthermore, the inefficient neuroinvasion in aged individuals may lead to significant levels of subclinical TSE disease in the population.
Collapse
Affiliation(s)
- Karen L Brown
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Roslin, United Kingdom
| | | | | | | | | |
Collapse
|
32
|
Sein Lwin, Inoshima Y, Ueno H, Ishiguro N. Uptake and transport of foreign particles in Peyer’s patches of both distal ileum and jejunum of calves. Cell Tissue Res 2009; 337:125-35. [DOI: 10.1007/s00441-009-0793-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 03/13/2009] [Indexed: 10/20/2022]
|
33
|
Mattei V, Barenco MG, Tasciotti V, Garofalo T, Longo A, Boller K, Löwer J, Misasi R, Montrasio F, Sorice M. Paracrine diffusion of PrP(C) and propagation of prion infectivity by plasma membrane-derived microvesicles. PLoS One 2009; 4:e5057. [PMID: 19337375 PMCID: PMC2659799 DOI: 10.1371/journal.pone.0005057] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Accepted: 02/23/2009] [Indexed: 11/18/2022] Open
Abstract
Cellular prion protein (PrPc) is a physiological constituent of eukaryotic cells. The cellular pathways underlying prions spread from the sites of prions infection/peripheral replication to the central nervous system are still not elucidated. Membrane-derived microvesicles (MVs) are submicron (0.1–1 µm) particles, that are released by cells during plasma membrane shedding processes. They are usually liberated from different cell types, mainly upon activation as well as apoptosis, in this case, one of their hallmarks is the exposure of phosphatidylserine in the outer leaflet of the membrane. MVs are also characterized by the presence of adhesion molecules, MHC I molecules, as well as of membrane antigens typical of their cell of origin. Evidence exists that MVs shedding provide vehicles to transfer molecules among cells, and that MVs are important modulators of cell-to-cell communication. In this study we therefore analyzed the potential role of membrane-derived MVs in the mechanism(s) of PrPC diffusion and prion infectivity transmission. We first identified PrPC in association with the lipid raft components Fyn, flotillin-2, GM1 and GM3 in MVs from plasma of healthy human donors. Similar findings were found in MVs from cell culture supernatants of murine neuronal cells. Furthermore we demonstrated that PrPSc is released from infected murine neuronal cells in association with plasma membrane-derived MVs and that PrPSc-bearing MVs are infectious both in vitro and in vivo. The data suggest that MVs may contribute both to the intercellular mechanism(s) of PrPC diffusion and signaling as well as to the process of prion spread and neuroinvasion.
Collapse
Affiliation(s)
- Vincenzo Mattei
- Dipartimento di Medicina Sperimentale, “Sapienza” University, Rome, Italy
- Laboratorio di Medicina Sperimentale e Patologia Ambientale, “Sapienza” University, Polo Universitario di Rieti “Sabina Universitas“, Rieti, Italy
| | - Maria Grazia Barenco
- Prion Research Group, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse, Langen, Germany
| | - Vincenzo Tasciotti
- Dipartimento di Medicina Sperimentale, “Sapienza” University, Rome, Italy
- Laboratorio di Medicina Sperimentale e Patologia Ambientale, “Sapienza” University, Polo Universitario di Rieti “Sabina Universitas“, Rieti, Italy
| | - Tina Garofalo
- Dipartimento di Medicina Sperimentale, “Sapienza” University, Rome, Italy
| | - Agostina Longo
- Dipartimento di Medicina Sperimentale, “Sapienza” University, Rome, Italy
| | - Klaus Boller
- Prion Research Group, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse, Langen, Germany
| | - Johannes Löwer
- Prion Research Group, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse, Langen, Germany
| | - Roberta Misasi
- Dipartimento di Medicina Sperimentale, “Sapienza” University, Rome, Italy
| | - Fabio Montrasio
- Prion Research Group, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse, Langen, Germany
| | - Maurizio Sorice
- Dipartimento di Medicina Sperimentale, “Sapienza” University, Rome, Italy
- Laboratorio di Medicina Sperimentale e Patologia Ambientale, “Sapienza” University, Polo Universitario di Rieti “Sabina Universitas“, Rieti, Italy
- * E-mail:
| |
Collapse
|
34
|
Cerovic V, McDonald V, Nassar MA, Paulin SM, Macpherson GG, Milling SWF. New insights into the roles of dendritic cells in intestinal immunity and tolerance. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 272:33-105. [PMID: 19121816 DOI: 10.1016/s1937-6448(08)01602-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) play a critical key role in the initiation of immune responses to pathogens. Paradoxically, they also prevent potentially damaging immune responses being directed against the multitude of harmless antigens, to which the body is exposed daily. These roles are particularly important in the intestine, where only a single layer of epithelial cells provides a barrier against billions of commensal microorganisms, pathogens, and food antigens, over a huge surface area. In the intestine, therefore, DCs are required to perform their dual roles very efficiently to protect the body from the dual threats of invading pathogens and unwanted inflammatory reactions. In this review, we first describe the biology of DCs and their interactions with other cells types, paying particular attention to intestinal DCs. We, then, examine the ways in which this biology may become misdirected, resulting in inflammatory bowel disease. Finally, we discuss how DCs potentiate immune responses against viral, bacterial, parasitic infections, and their importance in the pathogenesis of prion diseases. We, therefore, provide an overview of the complex cellular interactions that affect intestinal DCs and control the balance between immunity and tolerance.
Collapse
Affiliation(s)
- Vuk Cerovic
- Sir William Dunn School of Pathology, Oxford University, Oxford, United Kingdom
| | | | | | | | | | | |
Collapse
|
35
|
Accelerated prion replication in, but prolonged survival times of, prion-infected CXCR3-/- mice. J Virol 2008; 82:12464-71. [PMID: 18842729 DOI: 10.1128/jvi.01371-08] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Prion diseases have a significant inflammatory component. Glia activation, which is associated with increased production of cytokines and chemokines, may play an important role in disease development. Among the chemokines upregulated highly and early upregulated during scrapie infections are ligands of CXCR3. To gain more insight into the role of CXCR3 in a prion model, CXCR3-deficient (CXCR3(-/-)) mice were infected intracerebrally with scrapie strain 139A and characterized in comparison to similarly infected wild-type controls. CXCR3(-/-) mice showed significantly prolonged survival times of up to 30 days on average. Surprisingly, however, they displayed accelerated accumulation of misfolded proteinase K-resistant prion protein PrP(Sc) and 20 times higher infectious prion titers than wild-type mice at the asymptomatic stage of the disease, indicating that these PrP isoforms may not be critical determinants of survival times. As demonstrated by immunohistochemistry, Western blotting, and gene expression analysis, CXCR3-deficient animals develop an excessive astrocytosis. However, microglia activation is reduced. Quantitative analysis of gliosis-associated gene expression alterations demonstrated reduced mRNA levels for a number of proinflammatory factors in CXCR3(-/-) compared to wild-type mice, indicating a weaker inflammatory response in the knockout mice. Taken together, this murine prion model identifies CXCR3 as disease-modifying host factor and indicates that inflammatory glial responses may act in concert with PrP(Sc) in disease development. Moreover, the results indicate that targeting CXCR3 for treatment of prion infections could prolong survival times, but the results also raise the concern that impairment of microglial migration by ablation or inhibition of CXCR3 could result in increased accumulation of misfolded PrP(Sc).
Collapse
|
36
|
Raymond CR, Aucouturier P, Mabbott NA. In vivo depletion of CD11c+ cells impairs scrapie agent neuroinvasion from the intestine. THE JOURNAL OF IMMUNOLOGY 2008; 179:7758-66. [PMID: 18025222 DOI: 10.4049/jimmunol.179.11.7758] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Following oral exposure, some transmissible spongiform encephalopathy (TSE) agents accumulate first upon follicular dendritic cells (DCs) in the GALT. Studies in mice have shown that TSE agent accumulation in the GALT, in particular the Peyer's patches, is obligatory for the efficient transmission of disease to the brain. However, the mechanism through which TSE agents are initially conveyed from the gut lumen to the GALT is not known. Studies have implicated migratory hemopoietic DCs in this process, but direct demonstration of their involvement in vivo is lacking. In this study, we have investigated the contribution of CD11c(+) DCs in scrapie agent neuroinvasion through use of CD11c-diptheria toxin receptor-transgenic mice in which CD11c(+) DCs can be specifically and transiently depleted. Using two distinct scrapie agent strains (ME7 and 139A scrapie agents), we show that when CD11c(+) DCs were transiently depleted in the GALT and spleen before oral exposure, early agent accumulation in these tissues was blocked. In addition, CD11c(+) cell depletion reduced susceptibility to oral scrapie challenge indicating that TSE agent neuroinvasion from the GALT was impaired. In conclusion, these data demonstrate that migratory CD11c(+) DCs play a key role in the translocation of the scrapie agent from the gut lumen to the GALT from which neuroinvasion subsequently occurs.
Collapse
Affiliation(s)
- Claudine R Raymond
- Roslin Institute Neuropathogenesis Unit and Royal (Dick) School of Veterinary Sciences, Edinburgh, UK
| | | | | |
Collapse
|
37
|
Zhou XM, Xu GX, Zhao DM. In vitro effect of prion peptide PrP 106–126 on mouse macrophages: Possible role of macrophages in transport and proliferation for prion protein. Microb Pathog 2008; 44:129-34. [PMID: 17904794 DOI: 10.1016/j.micpath.2007.08.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Accepted: 08/06/2007] [Indexed: 11/20/2022]
Abstract
While there is a growing consensus on the understanding that the immune system plays an important role in facilitating the spread of prion infections from the periphery to the central nervous system, little is known about the key players in the first steps of the infection and about the sites of the disease development. Owing to their subepithelial location and their migratory capacity, macrophages could be early targets for prion transportation or propagation during the later stages of disease. In order to investigate the role of macrophages, we studied in vitro the effect of exposing primary peritoneal macrophages to a synthetic peptide homologous to residues 106-126 of the human prion protein (PrP), PrP 106-126. As shown by MTT assay, macrophage viability treated with less than 50 microM PrP 106-126 for 72 h was not inhibited but slightly stimulated at 10 and 25 microM, while there was significant decrease when exposed to 100 microM PrP 106-126 for 72 h. The expressions of PrP at mRNA and protein level were up-regulated following treatment with PrP 106-126 for 72 h. Cytokine TNF-alpha production were elevated by the PrP peptide in a time-dependent manner, which demonstrated a proinflammatory response linked to the presence and progression of prion disease took place in macrophages. These findings suggested that macrophages may play roles in the transportation and replication of the infectious agent.
Collapse
Affiliation(s)
- X M Zhou
- Animal Transmissible Spongiform Encephalopathies Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | | | | |
Collapse
|
38
|
Gilch S, Schmitz F, Aguib Y, Kehler C, Bülow S, Bauer S, Kremmer E, Schätzl HM. CpG and LPS can interfere negatively with prion clearance in macrophage and microglial cells. FEBS J 2007; 274:5834-44. [PMID: 17944938 DOI: 10.1111/j.1742-4658.2007.06105.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cells of the innate immune system play important roles in the progression of prion disease after peripheral infection. It has been found in vivo and in vitro that the expression of the cellular prion protein (PrP(c)) is up-regulated on stimulation of immune cells, also indicating the functional importance of PrP(c) in the immune system. The aim of our study was to investigate the impact of cytosine-phosphate-guanosine- and lipopolysaccharide-induced PrP(c) up-regulation on the uptake and processing of the pathological prion protein (PrP(Sc)) in phagocytic innate immune cells. For this purpose, we challenged the macrophage cell line J774, the microglial cell line BV-2 and primary bone marrow-derived macrophages in a resting or stimulated state with various prion strains, and monitored the uptake and clearance of PrP(Sc). Interestingly, stimulation led either to a transient increase in the level of PrP(Sc) relative to unstimulated cells or to a decelerated degradation of PrP(Sc). These features were dependent on cell type and prion strain. Our data indicate that the stimulation of innate immune cells may be able to support transient prion propagation, possibly explained by an increased PrP(c) cell surface expression in stimulated cells. We suggest that stimulation of innate immune cells can lead to an imbalance between the propagation and degradation of PrP(Sc).
Collapse
Affiliation(s)
- Sabine Gilch
- Institute of Virology, Prion Research Group, Technical University of Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Beekes M, McBride PA. The spread of prions through the body in naturally acquired transmissible spongiform encephalopathies. FEBS J 2007; 274:588-605. [PMID: 17288548 DOI: 10.1111/j.1742-4658.2007.05631.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Transmissible spongiform encephalopathies are fatal neurodegenerative diseases that are caused by unconventional pathogens and affect the central nervous system of animals and humans. Several different forms of these diseases result from natural infection (i.e. exposure to transmissible spongiform encephalopathy agents or prions, present in the natural environment of the respective host). This holds true also for scrapie in sheep, bovine spongiform encephalopathy in cattle, chronic wasting disease in elk and deer, or variant Creutzfeldt-Jakob disease in humans, all of which are assumed to originate predominantly from peroral prion infection. This article intends to provide an overview of the current state of knowledge on the spread of scrapie, chronic wasting disease, bovine spongiform encephalopathy and variant Creutzfeldt-Jakob disease agents through the body in naturally affected hosts, and in model animals experimentally challenged via the alimentary tract. Special attention is given to the tissue components and spreading pathways involved in the key stages of prion routing through the body, such as intestinal uptake, neuroinvasion of nerves and the central nervous system, and centrifugal spread from the brain and spinal cord to peripheral sites (e.g. sensory ganglia or muscles). The elucidation of the pathways and mechanisms by which prions invade a host and spread through the organism can contribute to efficient infection control strategies and the improvement of transmissible spongiform encephalopathy diagnostics. It may also help to identify prophylactic or therapeutic approaches that would impede naturally acquired transmissible spongiform encephalopathy infections.
Collapse
Affiliation(s)
- Michael Beekes
- Robert Koch-Institut (P24 - Transmissible Spongiforme Enzephalopathien), Berlin, Germany.
| | | |
Collapse
|
40
|
Glaysher BR, Mabbott NA. Role of the GALT in scrapie agent neuroinvasion from the intestine. THE JOURNAL OF IMMUNOLOGY 2007; 178:3757-66. [PMID: 17339474 DOI: 10.4049/jimmunol.178.6.3757] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Following oral exposure, some transmissible spongiform encephalopathy (TSE) agents accumulate first upon follicular dendritic cells (FDCs) in the GALT. Studies in mice have shown that this accumulation is obligatory for the efficient delivery of the TSE agent to the brain. However, which GALTs are crucial for disease pathogenesis is uncertain. Mice deficient in specific GALT components were used here to determine their separate involvement in scrapie agent neuroinvasion from the intestine. In the combined absence of the GALTs and FDCs (lymphotoxin (LT)alpha(-/-) mice and LTbeta(-/-) mice), scrapie agent transmission was blocked. When FDC maturation was induced in remaining lymphoid tissues, mice that lacked both Peyer's patches (PPs) and mesenteric lymph nodes (wild-type (WT)-->LTalpha(-/-) mice) or PPs alone (WT-->LTbeta(-/-) mice) remained refractory to disease, demonstrating an important role for the PPs. Although early scrapie agent accumulation also occurs within the mesenteric lymph nodes, their presence in WT-->LTbeta(-/-) mice did not restore disease susceptibility. We have also shown that isolated lymphoid follicles (ILFs) are important novel sites of TSE agent accumulation in the intestine. Mice that lacked PPs but contained numerous FDC-containing mature ILFs succumbed to scrapie at similar times to control mice. Because the formation and maturation status of ILFs is inducible and influenced by the gut flora, our data suggest that such factors could dramatically affect susceptibility to orally acquired TSE agents. In conclusion, these data demonstrate that following oral exposure TSE agent accumulation upon FDCs within lymphoid tissue within the intestine itself is critically required for efficient neuroinvasion.
Collapse
Affiliation(s)
- Bridget R Glaysher
- Neuropathogenesis Unit, Institute for Animal Health, Ogston Building, West Mains Road, Edinburgh, United Kingdom
| | | |
Collapse
|
41
|
PRNP promoter polymorphisms are associated with BSE susceptibility in Swiss and German cattle. BMC Genet 2007; 8:15. [PMID: 17437640 PMCID: PMC1857697 DOI: 10.1186/1471-2156-8-15] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2006] [Accepted: 04/16/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Non-synonymous polymorphisms within the prion protein gene (PRNP) influence the susceptibility and incubation time for transmissible spongiform encephalopathies (TSE) in some species such as sheep and humans. In cattle, none of the known polymorphisms within the PRNP coding region has a major influence on susceptibility to bovine spongiform encephalopathy (BSE). Recently, however, we demonstrated an association between susceptibility to BSE and a 23 bp insertion/deletion (indel) polymorphism and a 12 bp indel polymorphism within the putative PRNP promoter region using 43 German BSE cases and 48 German control cattle. The objective of this study was to extend this work by including a larger number of BSE cases and control cattle of German and Swiss origin. RESULTS Allele, genotype and haplotype frequencies of the two indel polymorphisms were determined in 449 BSE cattle and 431 unaffected cattle from Switzerland and Germany including all 43 German BSE and 16 German control animals from the original study. When breeds with similar allele and genotype distributions were compared, the 23 bp indel polymorphism again showed a significant association with susceptibility to BSE. However, some additional breed-specific allele and genotype distributions were identified, mainly related to the Brown breeds. CONCLUSION Our study corroborated earlier findings that polymorphisms in the PRNP promoter region have an influence on susceptibility to BSE. However, breed-specific differences exist that need to be accounted for when analyzing such data.
Collapse
|
42
|
Defaweux V, Dorban G, Antoine N, Piret J, Gabriel A, Jacqmot O, Falisse-Poirier N, Flandroy S, Zorzi D, Heinen E. Neuroimmune connections in jejunal and ileal Peyer’s patches at various bovine ages: potential sites for prion neuroinvasion. Cell Tissue Res 2007; 329:35-44. [PMID: 17406903 DOI: 10.1007/s00441-007-0396-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Accepted: 02/08/2007] [Indexed: 10/23/2022]
Abstract
During preclinical stages of cattle orally infected with bovine spongiform encephalopathy (BSE), the responsible agent is confined to ileal Peyer's patches (IPP), namely in nerve fibers and in lymph follicles, before reaching the peripheral and central nervous systems. No infectivity has been reported in other bovine lymphoid organs, including jejunal Peyer's patches (JPP). To determine the potential sites for prion neuroinvasion in IPP, we analyzed the mucosal innervation and the interface between nerve fibers and follicular dendritic cells (FDC), two dramatic influences on neuroinvasion. Bovine IPP were studied at three ages, viz., newborn calves, calves less than 12 months old, and bovines older than 24 months, and the parameters obtained were compared with those of JPP. No differences in innervation patterns between IPP and JPP were found. The major difference observed was that, in calves of less than 12 months, IPP were the major mucosal-associated lymphoid organ that possessed a large number of follicles with extended FDC networks. Using a panel of antibodies, we showed that PP in 24-month-old bovines were highly innervated at various strategic sites assumed to be involved in the invasion and replication of the BSE pathogen: the suprafollicular dome, T cell area, and germinal centers. In PP in calves of less than 12 months old, no nerve fibers positive for the neurofilament markers NF-L (70 kDa) and NF-H (200 kDa) were observed in contact with FDC. Thus, in view of the proportion of these protein subunits present in neurofilaments, the innervation of the germinal centers can be said to be an age-dependent dynamic process. This variation in innervation might influence the path of neuroinvasion and, thus, the susceptibility of bovines to the BSE agent.
Collapse
Affiliation(s)
- Valérie Defaweux
- Institute of Human Histology, Department of Morphology and Immunology, Faculty of Medicine, University of Liège, Liège, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Martínez del Hoyo G, López-Bravo M, Metharom P, Ardavín C, Aucouturier P. Prion Protein Expression by Mouse Dendritic Cells Is Restricted to the Nonplasmacytoid Subsets and Correlates with the Maturation State. THE JOURNAL OF IMMUNOLOGY 2006; 177:6137-42. [PMID: 17056541 DOI: 10.4049/jimmunol.177.9.6137] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Expression of the physiological cellular prion protein (PrP(C)) is remarkably regulated during differentiation and activation of cells of the immune system. Among these, dendritic cells (DCs) display particularly high levels of membrane PrP(C), which increase upon maturation, in parallel with that of molecules involved in Ag presentation to T cells. Freshly isolated mouse Langerhans cells, dermal DCs, and DCs from thymus, spleen, and mesenteric lymph nodes expressed low to intermediate levels of PrP(C). Highest levels of both PrP(C) and MHC class II molecules were displayed by lymph node CD8alpha(int) DCs, which represent fully mature cells having migrated from peripheral tissues. Maturation induced by overnight culture resulted in increased levels of surface PrP(C), as did in vivo DC activation by bacterial LPS. Studies on Fms-like tyrosine kinase 3 ligand bone marrow-differentiated B220(-) DCs confirmed that PrP(C) expression followed that of MHC class II and costimulatory molecules, and correlated with IL-12 production in response to TLR-9 engagement by CpG. However, at variance with conventional DCs, B220(+) plasmacytoid DCs isolated from the spleen, or in vitro differentiated, did not significantly express PrP(C), both before and after activation by TLR-9 engagement. PrP knockout mice displayed higher numbers of spleen CD8alpha(+) DCs, but no significant differences in their maturation response to stimulation through TLR-4 and TLR-9 were noticed. Results are discussed in relation to the functional relevance of PrP(C) expression by DCs in the induction of T cell responses, and to the pathophysiology of prion diseases.
Collapse
Affiliation(s)
- Gloria Martínez del Hoyo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Cientificas, Universidad Autónoma, Madrid, Spain
| | | | | | | | | |
Collapse
|
44
|
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
|
45
|
Abstract
Prion diseases are neurodegenerative conditions that cause extensive damage to nerve cells within the brain and can be fatal. Some prion disease agents accumulate first in lymphoid tissues, as they make their journey from the site of infection, such as the gut, to the brain. Studies in mouse models have shown that this accumulation is obligatory for the efficient delivery of prions to the brain. Indeed, if the accumulation of prions in lymphoid tissues is blocked, disease susceptibility is reduced. Therefore, the identification of the cells and molecules that are involved in the delivery of prions to the brain might identify targets for therapeutic intervention. This review describes the current understanding of the mechanisms involved in the delivery of prions to the brain.
Collapse
Affiliation(s)
- Neil A Mabbott
- Institute for Animal Health, Ogston Building, West Mains Road, Edinburgh EH9 3JF, UK.
| | | |
Collapse
|