1
|
Guijarro IM, Garcés M, Badiola JJ, Monzón M. In situ assessment of neuroinflammatory cytokines in different stages of ovine natural prion disease. Front Vet Sci 2024; 11:1404770. [PMID: 39493812 PMCID: PMC11528339 DOI: 10.3389/fvets.2024.1404770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/18/2024] [Indexed: 11/05/2024] Open
Abstract
Introduction According to the neuroinflammatory hypothesis, a cytokine-mediated host innate immune response may be involved in the mechanisms that contribute to the process of neurodegeneration. Specifically, regarding prion diseases, some experimental murine models have evidenced an altered profile of inflammatory intermediaries. However, the local inflammatory response has rarely been assessed, and never in tissues from different natural models throughout the progression of neurodegeneration. Methods The aim of this study was to use immunohistochemistry (IHC) to in situ assess the temporal protein expression of several cytokines in the cerebellum of sheep suffering from various clinical stages of scrapie. Results and discussion Clear changes in the expression of most of the assessed markers were observed in the affected sheep compared to the healthy control sheep, and from different stages. In summary, this preliminary IHC study focusing in the Purkinje cell layer changes demonstrate that all cytokines or respective receptors studied (IL-1, IL-1R, IL-2R, IL-6, IL-10R, and TNFαR) except for IFNγR are disease-associated signaling proteins showing an increase or decrease in relation to the progression of clinical disease. In the future, this study will be extended to other inflammatory mediators and brain regions, focusing in particular on the release of these inflammatory mediators by astroglial and microglial populations.
Collapse
Affiliation(s)
| | | | | | - Marta Monzón
- Research Centre for Encephalopathies and Transmissible Emerging Diseases. Institute for Health Research Aragón (IIS) – WOAH Reference Laboratory for BSE and Scrapie, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
2
|
Assis-de-Lemos G, Moura-do-Nascimento R, Amaral-do-Nascimento M, Miceli AC, Vieira TCRG. Interactions between Cytokines and the Pathogenesis of Prion Diseases: Insights and Implications. Brain Sci 2024; 14:413. [PMID: 38790392 PMCID: PMC11117815 DOI: 10.3390/brainsci14050413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/16/2024] [Accepted: 04/21/2024] [Indexed: 05/26/2024] Open
Abstract
Transmissible Spongiform Encephalopathies (TSEs), including prion diseases such as Bovine Spongiform Encephalopathy (Mad Cow Disease) and variant Creutzfeldt-Jakob Disease, pose unique challenges to the scientific and medical communities due to their infectious nature, neurodegenerative effects, and the absence of a cure. Central to the progression of TSEs is the conversion of the normal cellular prion protein (PrPC) into its infectious scrapie form (PrPSc), leading to neurodegeneration through a complex interplay involving the immune system. This review elucidates the current understanding of the immune response in prion diseases, emphasizing the dual role of the immune system in both propagating and mitigating the disease through mechanisms such as glial activation, cytokine release, and blood-brain barrier dynamics. We highlight the differential cytokine profiles associated with various prion strains and stages of disease, pointing towards the potential for cytokines as biomarkers and therapeutic targets. Immunomodulatory strategies are discussed as promising avenues for mitigating neuroinflammation and delaying disease progression. This comprehensive examination of the immune response in TSEs not only advances our understanding of these enigmatic diseases but also sheds light on broader neuroinflammatory processes, offering hope for future therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | - Tuane C. R. G. Vieira
- Institute of Medical Biochemistry Leopoldo de Meis and National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.A.-d.-L.); (R.M.-d.-N.); (M.A.-d.-N.); (A.C.M.)
| |
Collapse
|
3
|
Shim KH, Sharma N, An SSA. Prion therapeutics: Lessons from the past. Prion 2022; 16:265-294. [PMID: 36515657 PMCID: PMC9754114 DOI: 10.1080/19336896.2022.2153551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 12/15/2022] Open
Abstract
Prion diseases are a group of incurable zoonotic neurodegenerative diseases (NDDs) in humans and other animals caused by the prion proteins. The abnormal folding and aggregation of the soluble cellular prion proteins (PrPC) into scrapie isoform (PrPSc) in the Central nervous system (CNS) resulted in brain damage and other neurological symptoms. Different therapeutic approaches, including stalling PrPC to PrPSc conversion, increasing PrPSc removal, and PrPC stabilization, for which a spectrum of compounds, ranging from organic compounds to antibodies, have been explored. Additionally, a non-PrP targeted drug strategy using serpin inhibitors has been discussed. Despite numerous scaffolds being screened for anti-prion activity in vitro, only a few were effective in vivo and unfortunately, almost none of them proved effective in the clinical studies, most likely due to toxicity and lack of permeability. Recently, encouraging results from a prion-protein monoclonal antibody, PRN100, were presented in the first human trial on CJD patients, which gives a hope for better future for the discovery of other new molecules to treat prion diseases. In this comprehensive review, we have re-visited the history and discussed various classes of anti-prion agents, their structure, mode of action, and toxicity. Understanding pathogenesis would be vital for developing future treatments for prion diseases. Based on the outcomes of existing therapies, new anti-prion agents could be identified/synthesized/designed with reduced toxicity and increased bioavailability, which could probably be effective in treating prion diseases.
Collapse
Affiliation(s)
- Kyu Hwan Shim
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Niti Sharma
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| |
Collapse
|
4
|
Tahir W, Thapa S, Schatzl HM. Astrocyte in prion disease: a double-edged sword. Neural Regen Res 2022; 17:1659-1665. [PMID: 35017412 PMCID: PMC8820723 DOI: 10.4103/1673-5374.332202] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/08/2021] [Accepted: 07/22/2021] [Indexed: 11/04/2022] Open
Abstract
Prion diseases are infectious protein misfolding disorders of the central nervous system that result from misfolding of the cellular prion protein (PrPC) into the pathologic isoform PrPSc. Pathologic hallmarks of prion disease are depositions of pathological prion protein PrPSc, neuronal loss, spongiform degeneration and astrogliosis in the brain. Prion diseases affect human and animals, there is no effective therapy, and they invariably remain fatal. For a long time, neuronal loss was considered the sole reason for neurodegeneration in prion pathogenesis, and the contribution of non-neuronal cells like microglia and astrocytes was considered less important. Recent evidence suggests that neurodegeneration during prion pathogenesis is a consequence of a complex interplay between neuronal and non-neuronal cells in the brain, but the exact role of these non-neuronal cells during prion pathology is still elusive. Astrocytes are non-neuronal cells that regulate brain homeostasis under physiological conditions. However, astrocytes can deposit PrPSc aggregates and propagate prions in prion-infected brains. Additionally, sub-populations of reactive astrocytes that include neurotrophic and neurotoxic species have been identified, differentially expressed in the brain during prion infection. Revealing the exact role of astrocytes in prion disease is hampered by the lack of in vitro models of prion-infected astrocytes. Recently, we established a murine astrocyte cell line persistently infected with mouse-adapted prions, and showed how such astrocytes differentially process various prion strains. Considering the complexity of the role of astrocytes in prion pathogenesis, we need more in vitro and in vivo models for exploring the contribution of sub-populations of reactive astrocytes, their differential regulation of signaling cascades, and the interaction with neurons and microglia during prion pathogenesis. This will help to establish novel in vivo models and define new therapeutic targets against prion diseases. In this review, we will discuss the complex role of astrocytes in prion disease, the existing experimental resources, the challenges to analyze the contribution of astrocytes in prion disease pathogenesis, and future strategies to improve the understanding of their role in prion disease.
Collapse
Affiliation(s)
- Waqas Tahir
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Calgary Prion Research Unit, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Simrika Thapa
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Calgary Prion Research Unit, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Hermann M. Schatzl
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Calgary Prion Research Unit, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
5
|
de Melo ASLF, Lima JLD, Malta MCS, Marroquim NF, Moreira ÁR, de Almeida Ladeia I, dos Santos Cardoso F, Gonçalves DB, Dutra BG, dos Santos JCC. The role of microglia in prion diseases and possible therapeutic targets: a literature review. Prion 2021; 15:191-206. [PMID: 34751640 PMCID: PMC8583147 DOI: 10.1080/19336896.2021.1991771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 10/01/2021] [Indexed: 11/19/2022] Open
Abstract
Creutzfeldt-Jakob disease (CJD) is a rare and fatal condition that leads to progressive neurodegeneration due to gliosis, vacuolation of central nervous system tissue, and loss of neurons. Microglia play a crucial role in maintaining Central Nervous System (CNS) homoeostasis, both in health and disease, through phagocytosis and cytokine production. In the context of CJD, the immunomodulatory function of microglia turns it into a cell of particular interest. Microglia would be activated by infectious prion proteins, initially acquiring a phagocytic and anti-inflammatory profile (M2), and producing cytokines such as IL-4, IL-10, and TGF-β. Therefore, microglia are seen as a key target for the development of new treatment approaches, with many emerging strategies to guide it towards a beneficial role upon neuroinflammation, by manipulating its metabolic pathways. In such a setting, many cellular targets in microglia that can be involved in phenotype modulation, such as membrane receptors, have been identified and pointed out as possible targets for further experiments and therapeutic approaches. In this article, we review the major findings about the role of microglia in CJD, including its relationship to some risk factors associated with the development of the disease. Furthermore, considering its central role in neural immunity, we explore microglial connection with other elements of the immune system and cell signalling, such as inflammasomes, the complement and purinergic systems, and the latest finding strategies to guide these cells from harmful to beneficial roles.
Collapse
Affiliation(s)
| | | | | | | | - Álvaro Rivelli Moreira
- Department of Neurology, Centro Universitário Governador Ozanam Coelho, UniFacog, Ubá, MG, Brazil
| | | | - Fabrizio dos Santos Cardoso
- Núcleo de Pesquisas Tecnológicas, Universidade De Mogi Das Cruzes, Mogi das Cruzes, SP, Brazil
- Department of Psychology and Institute for Neuroscience, University of Texas (Ut), Austin, TX, USA
| | | | | | - Júlio César Claudino dos Santos
- Laboratório de Neurociências, Departamento De Neurologia E Neurocirurgia, Universidade Federal de São Paulo, São Paulo, Sp, Brazil
| |
Collapse
|
6
|
Neuroimmune Response Mediated by Cytokines in Natural Scrapie after Chronic Dexamethasone Treatment. Biomolecules 2021; 11:biom11020204. [PMID: 33540568 PMCID: PMC7912810 DOI: 10.3390/biom11020204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 01/27/2021] [Indexed: 01/15/2023] Open
Abstract
The actual role of prion protein-induced glial activation and subsequent cytokine secretion during prion diseases is still incompletely understood. The overall aim of this study is to assess the effect of an anti-inflammatory treatment with dexamethasone on different cytokines released by neuroglial cells that are potentially related to neuroinflammation in natural scrapie. This study emphasizes the complex interactions existent among several pleiotropic neuromodulator peptides and provides a global approach to clarify neuroinflammatory processes in prion diseases. Additionally, an impairment of communication between microglial and astroglial populations mediated by cytokines, mainly IL-1, is suggested. The main novelty of this study is that it is the first one assessing in situ neuroinflammatory activity in relation to chronic anti-inflammatory therapy, gaining relevance because it is based on a natural model. The cytokine profile data would suggest the activation of some neurotoxicity-associated route. Consequently, targeting such a pathway might be a new approach to modify the damaging effects of neuroinflammation.
Collapse
|
7
|
Guijarro IM, Garcés M, Marín B, Otero A, Barrio T, Badiola JJ, Monzón M. Neuroimmune Response in Natural Preclinical Scrapie after Dexamethasone Treatment. Int J Mol Sci 2020; 21:ijms21165779. [PMID: 32806582 PMCID: PMC7460817 DOI: 10.3390/ijms21165779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/04/2020] [Accepted: 08/10/2020] [Indexed: 12/18/2022] Open
Abstract
A recently published report on chronic dexamethasone treatment for natural scrapie supported the hypothesis of the potential failure of astroglia in the advanced stage of disease. Herein, we aimed to extend the aforementioned study on the effect of this anti-inflammatory therapy to the initial phase of scrapie, with the aim of elucidating the natural neuroinflammatory process occurring in this neurodegenerative disorder. The administration of this glucocorticoid resulted in an outstanding reduction in vacuolation and aberrant protein deposition (nearly null), and an increase in glial activation. Furthermore, evident suppression of IL-1R and IL-6 and the exacerbation of IL-1α, IL-2R, IL-10R and IFNγR were also demonstrated. Consequently, the early stage of the disease is characterized by an intact neuroglial response similar to that of healthy individuals attempting to re-establish homeostasis. A complex network of neuroinflammatory markers is involved from the very early stages of this prion disease, which probably becomes impaired in the more advanced stages. The in vivo animal model used herein provides essential observations on the pathogenesis of natural scrapie, as well as the possibility of establishing neuroglia as potential target cells for anti-inflammatory therapy.
Collapse
|
8
|
Sheikh MH, Solito E. Annexin A1: Uncovering the Many Talents of an Old Protein. Int J Mol Sci 2018; 19:E1045. [PMID: 29614751 PMCID: PMC5979524 DOI: 10.3390/ijms19041045] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 03/07/2018] [Accepted: 03/15/2018] [Indexed: 12/11/2022] Open
Abstract
Annexin A1 (ANXA1) has long been classed as an anti-inflammatory protein due to its control over leukocyte-mediated immune responses. However, it is now recognized that ANXA1 has widespread effects beyond the immune system with implications in maintaining the homeostatic environment within the entire body due to its ability to affect cellular signalling, hormonal secretion, foetal development, the aging process and development of disease. In this review, we aim to provide a global overview of the role of ANXA1 covering aspects of peripheral and central inflammation, immune repair and endocrine control with focus on the prognostic, diagnostic and therapeutic potential of the molecule in cancer, neurodegeneration and inflammatory-based disorders.
Collapse
Affiliation(s)
- Madeeha H Sheikh
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Egle Solito
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|
9
|
Ragagnin A, Ezpeleta J, Guillemain A, Boudet-Devaud F, Haeberlé AM, Demais V, Vidal C, Demuth S, Béringue V, Kellermann O, Schneider B, Grant NJ, Bailly Y. Cerebellar compartmentation of prion pathogenesis. Brain Pathol 2017; 28:240-263. [PMID: 28268246 DOI: 10.1111/bpa.12503] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/01/2017] [Indexed: 02/06/2023] Open
Abstract
In prion diseases, the brain lesion profile is influenced by the prion "strain" properties, the invasion route to the brain, and still unknown host cell-specific parameters. To gain insight into those endogenous factors, we analyzed the histopathological alterations induced by distinct prion strains in the mouse cerebellum. We show that 22L and ME7 scrapie prion proteins (PrP22L , PrPME7 ), but not bovine spongiform encephalopathy PrP6PB1 , accumulate in a reproducible parasagittal banding pattern in the cerebellar cortex of infected mice. Such banding pattern of PrP22L aggregation did not depend on the neuroinvasion route, but coincided with the parasagittal compartmentation of the cerebellum mostly defined by the expression of zebrins, such as aldolase C and the excitatory amino acid transporter 4, in Purkinje cells. We provide evidence that Purkinje cells display a differential, subtype-specific vulnerability to 22L prions with zebrin-expressing Purkinje cells being more resistant to prion toxicity, while in stripes where PrP22L accumulated most zebrin-deficient Purkinje cells are lost and spongiosis accentuated. In addition, in PrP22L stripes, enhanced reactive astrocyte processes associated with microglia activation support interdependent events between the topographic pattern of Purkinje cell death, reactive gliosis and PrP22L accumulation. Finally, we find that in preclinically-ill mice prion infection promotes at the membrane of astrocytes enveloping Purkinje cell excitatory synapses, upregulation of tumor necrosis factor-α receptor type 1 (TNFR1), a key mediator of the neuroinflammation process. These overall data show that Purkinje cell sensitivity to prion insult is locally restricted by the parasagittal compartmentation of the cerebellum, and that perisynaptic astrocytes may contribute to prion pathogenesis through prion-induced TNFR1 upregulation.
Collapse
Affiliation(s)
- Audrey Ragagnin
- Cytologie et Cytopathologie Neuronales, Institut des Neurosciences Cellulaires & Intégratives, CNRS UPR 3212, Strasbourg, France
| | - Juliette Ezpeleta
- INSERM UMR-S1124, Cellules Souches, Signalisation et Prions, Université Paris Descartes, Paris, France
| | - Aurélie Guillemain
- Cytologie et Cytopathologie Neuronales, Institut des Neurosciences Cellulaires & Intégratives, CNRS UPR 3212, Strasbourg, France
| | - François Boudet-Devaud
- INSERM UMR-S1124, Cellules Souches, Signalisation et Prions, Université Paris Descartes, Paris, France
| | - Anne-Marie Haeberlé
- Cytologie et Cytopathologie Neuronales, Institut des Neurosciences Cellulaires & Intégratives, CNRS UPR 3212, Strasbourg, France
| | - Valérie Demais
- Plateforme Imagerie In Vitro, CNRS UPS-3156, Université de Strasbourg, Strasbourg, France
| | | | - Stanislas Demuth
- Cytologie et Cytopathologie Neuronales, Institut des Neurosciences Cellulaires & Intégratives, CNRS UPR 3212, Strasbourg, France
| | | | - Odile Kellermann
- INSERM UMR-S1124, Cellules Souches, Signalisation et Prions, Université Paris Descartes, Paris, France
| | - Benoit Schneider
- INSERM UMR-S1124, Cellules Souches, Signalisation et Prions, Université Paris Descartes, Paris, France
| | - Nancy J Grant
- Cytologie et Cytopathologie Neuronales, Institut des Neurosciences Cellulaires & Intégratives, CNRS UPR 3212, Strasbourg, France
| | - Yannick Bailly
- Cytologie et Cytopathologie Neuronales, Institut des Neurosciences Cellulaires & Intégratives, CNRS UPR 3212, Strasbourg, France
| |
Collapse
|
10
|
Brandel JP, Haïk S. Malattie da prioni o encefalopatie spongiformi trasmissibili. Neurologia 2016. [DOI: 10.1016/s1634-7072(16)77562-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
11
|
Nuvolone M, Sorce S, Schwarz P, Aguzzi A. Prion pathogenesis in the absence of NLRP3/ASC inflammasomes. PLoS One 2015; 10:e0117208. [PMID: 25671600 PMCID: PMC4324825 DOI: 10.1371/journal.pone.0117208] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/19/2014] [Indexed: 12/31/2022] Open
Abstract
The accumulation of the scrapie prion protein PrPSc, a misfolded conformer of the cellular prion protein PrPC, is a crucial feature of prion diseases. In the central nervous system, this process is accompanied by conspicuous microglia activation. The NLRP3 inflammasome is a multi-molecular complex which can sense heterogeneous pathogen-associated molecular patterns and culminates in the activation of caspase 1 and release of IL 1β. The NLRP3 inflammasome was reported to be essential for IL 1β release after in vitro exposure to the amyloidogenic peptide PrP106-126 and to recombinant PrP fibrils. We therefore studied the role of the NLRP3 inflammasome in a mouse model of prion infection. Upon intracerebral inoculation with scrapie prions (strain RML), mice lacking NLRP3 (Nlrp3-/-) or the inflammasome adaptor protein ASC (Pycard-/-) succumbed to scrapie with attack rates and incubation times similar to wild-type mice, and developed the classic histologic and biochemical features of prion diseases. Genetic ablation of NLRP3 or ASC did not significantly impact on brain levels of IL 1β at the terminal stage of disease. Our results exclude a significant role for NLRP3 and ASC in prion pathogenesis and invalidate their claimed potential as therapeutic target against prion diseases.
Collapse
Affiliation(s)
- Mario Nuvolone
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
- * E-mail: (AA); (MN)
| | - Silvia Sorce
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Petra Schwarz
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
- * E-mail: (AA); (MN)
| |
Collapse
|
12
|
Pathogenic prion protein fragment (PrP106-126) promotes human immunodeficiency virus type-1 infection in peripheral blood monocyte-derived macrophages. Virology 2015; 476:372-376. [PMID: 25589240 DOI: 10.1016/j.virol.2014.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/11/2014] [Accepted: 11/16/2014] [Indexed: 11/22/2022]
Abstract
Transfusion of blood and blood products contaminated with the pathogenic form of prion protein Prp(sc), thought to be the causative agent of variant a Creutzfeldt-Jakob disease (vCJD), may result in serious consequences in recipients with a compromised immune system, for example, as seen in HIV-1 infection. In the present study, we demonstrate that treatment of peripheral blood monocyte-derived macrophages (MDM) with PrP106-126, a synthetic domain of PrP(sc) that has intrinsic functional activities related to the full-length protein, markedly increased their susceptibility to HIV-1 infection, induced cytokine secretion, and enhanced their migratory behavior in response to N-formyl-l-methionyl-l-leucyl-l-phenylalanine (fMLP). Live-cell imaging of MDM cultured in the presence of PrP106-126 showed large cell clusters indicative of cellular activation. Tyrosine kinase inhibitor STI-571, protein kinase C inhibitor K252B, and cyclin-dependent kinase inhibitor olomoucine attenuated PrP106-126-induced altered MDM functions. These findings delineate a previously undefined functional role of PrP106-126-mediated host cell response in promoting HIV-1 pathogenesis.
Collapse
|
13
|
Prion infection of mouse brain reveals multiple new upregulated genes involved in neuroinflammation or signal transduction. J Virol 2014; 89:2388-404. [PMID: 25505076 DOI: 10.1128/jvi.02952-14] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED Gliosis is often a preclinical pathological finding in neurodegenerative diseases, including prion diseases, but the mechanisms facilitating gliosis and neuronal damage in these diseases are not understood. To expand our knowledge of the neuroinflammatory response in prion diseases, we assessed the expression of key genes and proteins involved in the inflammatory response and signal transduction in mouse brain at various times after scrapie infection. In brains of scrapie-infected mice at pre- and postclinical stages, we identified 15 previously unreported differentially expressed genes related to inflammation or activation of the STAT signal transduction pathway. Levels for the majority of differentially expressed genes increased with time postinfection. In quantitative immunoblotting experiments of STAT proteins, STAT1α, phosphorylated-STAT1α (pSTAT1α), and pSTAT3 were increased between 94 and 131 days postinfection (p.i.) in brains of mice infected with strain 22L. Furthermore, a select group of STAT-associated genes was increased preclinically during scrapie infection, suggesting early activation of the STAT signal transduction pathway. Comparison of inflammatory markers between mice infected with scrapie strains 22L and RML indicated that the inflammatory responses and gene expression profiles in the brains were strikingly similar, even though these scrapie strains infect different brain regions. The endogenous interleukin-1 receptor antagonist (IL-1Ra), an inflammatory marker, was newly identified as increasing preclinically in our model and therefore might influence scrapie pathogenesis in vivo. However, in IL-1Ra-deficient or overexpressor transgenic mice inoculated with scrapie, neither loss nor overexpression of IL-1Ra demonstrated any observable effect on gliosis, protease-resistant prion protein (PrPres) formation, disease tempo, pathology, or expression of the inflammatory genes analyzed. IMPORTANCE Prion infection leads to PrPres deposition, gliosis, and neuroinflammation in the central nervous system before signs of clinical illness. Using a scrapie mouse model of prion disease to assess various time points postinoculation, we identified 15 unreported genes that were increased in the brains of scrapie-infected mice and were associated with inflammation and/or JAK-STAT activation. Comparison of mice infected with two scrapie strains (22L and RML), which have dissimilar neuropathologies, indicated that the inflammatory responses and gene expression profiles in the brains were similar. Genes that increased prior to clinical signs might be involved in controlling scrapie infection or in facilitating damage to host tissues. We tested the possible role of the endogenous IL-1Ra, which was increased at 70 days p.i. In scrapie-infected mice deficient in or overexpressing IL-1Ra, there was no observable effect on gliosis, PrPres formation, disease tempo, pathology, or expression of inflammatory genes analyzed.
Collapse
|
14
|
Motoyoshi-Yamashiro A, Takano K, Kawabe K, Izawa T, Nakajima H, Moriyama M, Nakamura Y. Amphotericin B induces glial cell line-derived neurotrophic factor in the rat brain. J Vet Med Sci 2014; 76:1353-8. [PMID: 25283947 PMCID: PMC4221168 DOI: 10.1292/jvms.14-0160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Amphotericin B (AmB) is a
polyene antifungal drug and is reported to be one of a few reagents having therapeutic
effects on prion diseases, that is, a delay in the appearance of clinical signs and
prolongation of the survival time in an animal model. In prion diseases, glial cells have
been suggested to play important roles; however, the therapeutic mechanism of AmB on prion
diseases remains elusive. We have previously reported that AmB changed the expression of
neurotrophic factors in microglia and astrocytes (Motoyoshi et al., 2008,
Neurochem. Int. 52, 1290–1296; Motoyoshi-Yamashiro et
al., 2013, ibid. 63, 93–100). These results suggested that
neurotrophic factors derived from glial cells might be involved in the therapeutic
mechanism of AmB. In the present study, we examined immunohistochemically the effects of
AmB on the expression of neurotrophic factors in the rat brain. We found that direct
injection of AmB into the striatum significantly enhanced the expression of glial cell
line-derived neurotrophic factor protein. Amphotericin B also increased the expressions of
CD11b and glial fibrillary acidic protein, markers of microglia and astrocytes,
respectively. Moreover, expressions of the two neurotrophic factors by AmB were
co-localized with the expression of CD11b or glial fibrillary acidic protein. These
results suggest that AmB in vivo might also activate glial cells and
induce the production of neurotrophic factors protecting neurons in prion diseases.
Collapse
Affiliation(s)
- Akiko Motoyoshi-Yamashiro
- Laboratory of Integrative Physiology in Veterinary Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka 598-8531, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Song K, Na JY, Oh MH, Kim S, Kim YH, Park BY, Shin GW, Kim B, You M, Kwon J. Synthetic prion Peptide 106-126 resulted in an increase matrix metalloproteinases and inflammatory cytokines from rat astrocytes and microglial cells. Toxicol Res 2013; 28:5-9. [PMID: 24278583 PMCID: PMC3834397 DOI: 10.5487/tr.2012.28.1.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/12/2012] [Accepted: 03/21/2012] [Indexed: 12/22/2022] Open
Abstract
It has been shown that the accumulation of prion in the cytoplasm can result in neurodegenerative disorders. Synthetic prion peptide 106-126 (PrP) is a glycoprotein that is expressed predominantly by neurons and other cells, including glial cells. Prion-induced chronic neurodegeneration has a substantial inflammatory component, and an increase in the levels of matrix metalloproteinases (MMPs) may play an important role in neurodegenerative development and progression. However, the expression of MMPs in PrP induced rat astrocytes and microglia has not yet been compared. Thus, in this study, we examined the fluorescence intensity of CD11b positive microglia and Glial Fibrillary Acidic Protein (GFAP) positive astrocytes and found that the fluorescent intensity was increased following incubation with PrP at 24 hours in a dose-dependent manner. We also observed an increase in interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) protein expression, which are initial inflammatory cytokines, in both PrP induced astrocytes and microglia. Furthermore, an increase MMP-1, 3 and 11 expressions in PrP induced astrocytes and microglia was observed by real time PCR. Our results demonstrated PrP induced activation of astrocytes and microglia respectively, which resulted in an increase in inflammatory cytokines and MMPs expression. These results provide the insight into the different sensitivities of glial cells to PrP.
Collapse
Affiliation(s)
- Kibbeum Song
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Jeonju 561-156, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
PDK1 decreases TACE-mediated α-secretase activity and promotes disease progression in prion and Alzheimer's diseases. Nat Med 2013; 19:1124-31. [DOI: 10.1038/nm.3302] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/16/2013] [Indexed: 12/17/2022]
|
17
|
Xie WL, Shi Q, Zhang J, Zhang BY, Gong HS, Guo Y, Wang SB, Xu Y, Wang K, Chen C, Liu Y, Dong XP. Abnormal activation of microglia accompanied with disrupted CX3CR1/CX3CL1 pathway in the brains of the hamsters infected with scrapie agent 263K. J Mol Neurosci 2013; 51:919-32. [PMID: 23526370 DOI: 10.1007/s12031-013-0002-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 03/11/2013] [Indexed: 10/27/2022]
Abstract
Microglial cells are resident mononuclear phagocytes of the central nervous system (CNS). Active proliferation of microglia in the brain has been identified in neurodegenerative disorders, including some kinds of prion disease. However, the detailed regional distribution between microglia and PrP(Sc) deposition has not been presented, and investigation of fractalkine signaling which is involved in the regulation of activation of microglia in prion disease is not well documented. In this study, the disease phenomenon of microglial accumulation in the CNS was thoroughly analyzed using a scrapie-infected experimental model. Western blots of microglia-specific markers Iba1 and CD68, immunohistochemical and immunofluorescent assays demonstrated obviously activation of microglia in almost whole brain regions in the infected animals. Under the dynamic analysis on hallmarks of activation of microglia, a time-dependent increase of Iba1 and CD68 was detected, accompanied by accumulation of PrP(Sc) and progression of neurodegenerative symptoms. With serial brain sections and double staining of Iba1 and PrP(Sc), we observed that the microglia distributed around PrP(Sc) deposits in 263K-infected hamsters' brains, proposing PrP(Sc) phagocytosis. Flow cytometry assays with the single-cell suspensions prepared from the cortical region of the infected brains verified an activation of microglial population. ELISA assays of the cytokines in brain homogenates revealed significant upregulations of interleukin (IL)-1β, IL-6 and TNF-α when infected. Evaluation of fractalkine signaling in the infected hamsters' brains showed progressively downregulation of CX3CL1 during the incubation. Prion peptide PrP106-126 also disrupted fractalkine and evoked microglial activation in rat primary neuron-glia mixed cultures. Our data here demonstrate an activated status of microglia in CNS tissues of infectious prion disease, possibly through fractalkine signaling deficiency.
Collapse
Affiliation(s)
- Wu-Ling Xie
- School of Medicine, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Corda E, Beck KE, Sallis RE, Vickery CM, Denyer M, Webb PR, Bellworthy SJ, Spencer YI, Simmons MM, Spiropoulos J. The interpretation of disease phenotypes to identify TSE strains in mice: characterisation of BSE using PrPSc distribution patterns in the brain. Vet Res 2012; 43:86. [PMID: 23245876 PMCID: PMC3567960 DOI: 10.1186/1297-9716-43-86] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 12/06/2012] [Indexed: 11/20/2022] Open
Abstract
In individual animals affected by transmissible spongiform encephalopathies, different disease phenotypes can be identified which are attributed to different strains of the agent. In the absence of reliable technology to fully characterise the agent, classification of disease phenotype has been used as a strain typing tool which can be applied in any host. This approach uses standardised data on biological parameters, established for a single host, to allow comparison of different prion sources. Traditionally prion strain characterisation in wild type mice is based on incubation periods and lesion profiles after the stabilisation of the agent into the new host which requires serial passages. Such analysis can take many years, due to prolonged incubation periods. The current study demonstrates that the PrPSc patterns produced by one serial passage in wild type mice of bovine or ovine BSE were consistent, stable and showed minimal and predictable differences from mouse-stabilised reference strains. This biological property makes PrPSc deposition pattern mapping a powerful tool in the identification and definition of TSE strains on primary isolation, making the process of characterisation faster and cheaper than a serial passage protocol. It can be applied to individual mice and therefore it is better suited to identify strain diversity within single inocula in case of co-infections or identify strains in cases where insufficient mice succumb to disease for robust lesion profiles to be constructed. The detailed description presented in this study provides a reference document for identifying BSE in wild type mice.
Collapse
Affiliation(s)
- Erica Corda
- School of Veterinary Medicine, University of Milan, Milan, Italy
| | - Katy E Beck
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB, United Kingdom
| | - Rosemary E Sallis
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB, United Kingdom
| | - Christopher M Vickery
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB, United Kingdom
| | - Margaret Denyer
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB, United Kingdom
| | - Paul R Webb
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB, United Kingdom
| | - Susan J Bellworthy
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB, United Kingdom
| | - Yvonne I Spencer
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB, United Kingdom
| | - Marion M Simmons
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB, United Kingdom
| | - John Spiropoulos
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB, United Kingdom
| |
Collapse
|
19
|
Last V, Williams A, Werling D. Inhibition of cytosolic Phospholipase A2 prevents prion peptide-induced neuronal damage and co-localisation with Beta III Tubulin. BMC Neurosci 2012; 13:106. [PMID: 22928663 PMCID: PMC3496594 DOI: 10.1186/1471-2202-13-106] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 08/21/2012] [Indexed: 11/10/2022] Open
Abstract
Background Activation of phospholipase A2 (PLA2) and the subsequent metabolism of arachidonic acid (AA) to prostaglandins have been shown to play an important role in neuronal death in neurodegenerative disease. Here we report the effects of the prion peptide fragment HuPrP106-126 on the PLA2 cascade in primary cortical neurons and translocation of cPLA2 to neurites. Results Exposure of primary cortical neurons to HuPrP106-126 increased the levels of phosphorylated cPLA2 and caused phosphorylated cPLA2 to relocate from the cell body to the cellular neurite in a PrP-dependent manner, a previously unreported observation. HuPrP106-126 also induced significant AA release, an indicator of cPLA2 activation; this preceded synapse damage and subsequent cellular death. The novel translocation of p-cPLA2 postulated the potential for exposure to HuPrP106-126 to result in a re-arrangement of the cellular cytoskeleton. However p-cPLA2 did not colocalise significantly with F-actin, intermediate filaments, or microtubule-associated proteins. Conversely, p-cPLA2 did significantly colocalise with the cytoskeletal protein beta III tubulin. Pre-treatment with the PLA2 inhibitor, palmitoyl trifluoromethyl ketone (PACOCF3) reduced cPLA2 activation, AA release and damage to the neuronal synapse. Furthermore, PACOCF3 reduced expression of p-cPLA2 in neurites and inhibited colocalisation with beta III tubulin, resulting in protection against PrP-induced cell death. Conclusions Collectively, these findings suggest that cPLA2 plays a vital role in the action of HuPrP106-126 and that the colocalisation of p-cPLA2 with beta III tubulin could be central to the progress of neurodegeneration caused by prion peptides. Further work is needed to define exactly how PLA2 inhibitors protect neurons from peptide-induced toxicity and how this relates to intracellular structural changes occurring in neurodegeneration.
Collapse
Affiliation(s)
- Victoria Last
- Department of Pathology and Infectious Diseases, Royal Veterinary College, Hawkshead Lane, North Mymms, Hertfordshire AL9 7TA, UK.
| | | | | |
Collapse
|
20
|
Stoeck K, Zerr I. Cellular immune activation markers neopterin and β2-microglobulin are not elevated in the cerebrospinal fluid of patients with Creutzfeldt-Jakob disease. J Neuroimmunol 2011; 233:228-32. [PMID: 21232804 DOI: 10.1016/j.jneuroim.2010.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 11/05/2010] [Accepted: 12/05/2010] [Indexed: 10/18/2022]
Abstract
In prion diseases, neuroimmunological responses include activation of microglia, astrocytosis and release of pro- and anti-inflammatory cytokines, which might substantially contribute to the neurodegenerative process. In this study we investigated neopterin and beta(β)2-microglobulin, as markers of cellular immune activation, in the cerebrospinal fluid (CSF) of patients with Creutzfeldt-Jakob disease (CJD) and of patients with other neurological and non-neurological diseases. CSF samples from CJD patients were collected in the framework of the German CJD Surveillance study. Concentrations of neopterin and β2-microglobulin were determined in CSF using ELISA. We could not obtain significant changes in CSF levels of neopterin and β2-microglobulin in CJD patients when compared to other neurological and non-neurological controls. In a subanalysis of CJD patients only, we could find significant elevated neopterin levels in patients with MV genotype, potentially reflecting a distinct disease pathology. Since autoimmune inflammatory disorders are important differential diagnoses in CJD, additional biomarker might be helpful in clinical setting.
Collapse
Affiliation(s)
- Katharina Stoeck
- Department of Neurology, University of Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | | |
Collapse
|
21
|
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
|
22
|
Phospholipase A2 inhibitors protect against prion and Abeta mediated synapse degeneration. Mol Neurodegener 2010; 5:13. [PMID: 20374666 PMCID: PMC2865460 DOI: 10.1186/1750-1326-5-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 04/08/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND An early event in the neuropathology of prion and Alzheimer's diseases is the loss of synapses and a corresponding reduction in the level of synaptophysin, a pre-synaptic membrane protein essential for neurotransmission. The molecular mechanisms involved in synapse degeneration in these diseases are poorly understood. In this study the process of synapse degeneration was investigated by measuring the synaptophysin content of cultured neurones incubated with the prion derived peptide (PrP82-146) or with Abeta1-42, a peptide thought to trigger pathogenesis in Alzheimer's disease. A pharmacological approach was used to screen cell signalling pathways involved in synapse degeneration. RESULTS Pre-treatment with phospholipase A2 inhibitors (AACOCF3, MAFP and aristolochic acids) protected against synapse degeneration in cultured cortical and hippocampal neurones incubated with PrP82-146 or Abeta1-42. Synapse degeneration was also observed following the addition of a specific phospholipase A2 activating peptide (PLAP) and the addition of PrP82-146 or Abeta1-42 activated cytoplasmic phospholipase A2 within synapses. Activation of phospholipase A2 is the first step in the generation of platelet-activating factor (PAF) and PAF receptor antagonists (ginkgolide B, Hexa-PAF and CV6029) protected against synapse degeneration induced by PrP82-146, Abeta1-42 and PLAP. PAF facilitated the production of prostaglandin E2, which also caused synapse degeneration and pre-treatment with the prostanoid E receptor antagonist AH13205 protected against PrP82-146, Abeta1-42 and PAF induced synapse degeneration. CONCLUSIONS Our results are consistent with the hypothesis that PrP82-146 and Abeta1-42trigger abnormal activation of cytoplasmic phospholipase A2 resident within synapses, resulting in elevated levels of PAF and prostaglandin E2that cause synapse degeneration. Inhibitors of this pathway that can cross the blood brain barrier may protect against the synapse degeneration seen during Alzheimer's or prion diseases.
Collapse
|
23
|
Bate C, Tayebi M, Diomede L, Salmona M, Williams A. Glimepiride reduces the expression of PrPc, prevents PrPSc formation and protects against prion mediated neurotoxicity in cell lines. PLoS One 2009; 4:e8221. [PMID: 20011040 PMCID: PMC2784943 DOI: 10.1371/journal.pone.0008221] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 11/11/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND A hallmark of the prion diseases is the conversion of the host-encoded cellular prion protein (PrP(C)) into a disease related, alternatively folded isoform (PrP(Sc)). The accumulation of PrP(Sc) within the brain is associated with synapse loss and ultimately neuronal death. Novel therapeutics are desperately required to treat neurodegenerative diseases including the prion diseases. PRINCIPAL FINDINGS Treatment with glimepiride, a sulphonylurea approved for the treatment of diabetes mellitus, induced the release of PrP(C) from the surface of prion-infected neuronal cells. The cell surface is a site where PrP(C) molecules may be converted to PrP(Sc) and glimepiride treatment reduced PrP(Sc) formation in three prion infected neuronal cell lines (ScN2a, SMB and ScGT1 cells). Glimepiride also protected cortical and hippocampal neurones against the toxic effects of the prion-derived peptide PrP82-146. Glimepiride treatment significantly reduce both the amount of PrP82-146 that bound to neurones and PrP82-146 induced activation of cytoplasmic phospholipase A(2) (cPLA(2)) and the production of prostaglandin E(2) that is associated with neuronal injury in prion diseases. Our results are consistent with reports that glimepiride activates an endogenous glycosylphosphatidylinositol (GPI)-phospholipase C which reduced PrP(C) expression at the surface of neuronal cells. The effects of glimepiride were reproduced by treatment of cells with phosphatidylinositol-phospholipase C (PI-PLC) and were reversed by co-incubation with p-chloromercuriphenylsulphonate, an inhibitor of endogenous GPI-PLC. CONCLUSIONS Collectively, these results indicate that glimepiride may be a novel treatment to reduce PrP(Sc) formation and neuronal damage in prion diseases.
Collapse
Affiliation(s)
- Clive Bate
- Department of Pathology and Infectious Diseases, Royal Veterinary College, North Mymms, United Kingdom.
| | | | | | | | | |
Collapse
|
24
|
Loss of cerebellar granule neurons is associated with punctate but not with large focal deposits of prion protein in Creutzfeldt-Jakob disease. J Neuropathol Exp Neurol 2009; 68:892-901. [PMID: 19606064 DOI: 10.1097/nen.0b013e3181af7f23] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Whether aggregates of prion protein (PrP) reflect neurotoxicity or are neuroprotective in prion diseases is unclear. To address this question, we performed a clinicopathologic study of cerebellar granular neurons in 100 patients affected with sporadic Creutzfeldt-Jakob disease (CJD). There was significant loss of these neurons in the subset of cases with Val/Val genotype at PRNP Codon 129 and Molecular Isotype 2 of abnormal PrP (sporadic CJD-VV2) (n=32) compared with both the other CJD subtypes and to controls. Pathological PrP deposits of the punctate-type (synaptic-type) in this subgroup correlated with neuronal loss and proliferation of astrocytes and microglia. By contrast, the numbers of large deposits (5- to 50-microm-diameter) and numbers of amyloid plaques did not correlate with neuronal loss. These findings are consistent with the view that large aggregates may protect neurons by sequestering neurotoxic PrP oligomers, whereas punctate deposits may indicate the location of neuronal death processes in CJD.
Collapse
|
25
|
Analysis of protein levels of 24 cytokines in scrapie agent-infected brain and glial cell cultures from mice differing in prion protein expression levels. J Virol 2009; 83:11244-53. [PMID: 19710140 DOI: 10.1128/jvi.01413-09] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Activation of microglia and astroglia is seen in many neurodegenerative diseases including prion diseases. Activated glial cells produce cytokines as a protective response against certain pathogens and as part of the host inflammatory response to brain damage. In addition, cytokines might also exacerbate tissue damage initiated by other processes. In the present work using multiplex assays to analyze protein levels of 24 cytokines in scrapie agent-infected C57BL/10 mouse brains, we observed elevation of CCL2, CCL5, CXCL1, CXCL10, granulocyte-macrophage colony-stimulating factor (GM-CSF), gamma interferon (IFN-gamma), interleukin 1alpha (IL-1alpha), IL-1beta, IL-6, and IL-12p40. Scrapie agent-infected wild-type mice and transgenic mice expressing anchorless prion protein (PrP) had similar cytokine responses in spite of extensive differences in neuropathology. Therefore, these responses may be primarily a reaction to brain damage induced by prion infection rather than specific inducers of a particular type of pathology. To study the roles of astroglia and microglia in these cytokine responses, primary glial cultures were exposed to scrapie agent-infected brain homogenates. Microglia produced only IL-12p40 and CXCL10, whereas astroglia produced these cytokines plus CCL2, CCL3, CCL5, CXCL1, G-CSF, IL-1beta, IL-6, IL-12p70, and IL-13. Glial cytokine responses from wild-type mice and transgenic mice expressing anchorless PrP differed only slightly, but glia from PrP-null mice produced only IL-12p40, indicating that PrP expression was required for scrapie agent induction of other cytokines detected. The difference in cytokine response between microglia and astroglia correlated with 20-fold-higher levels of PrP expression in astroglia versus microglia, suggesting that high-level PrP expression on astroglia might be important for induction of certain cytokines.
Collapse
|
26
|
Abstract
Prions are infectious proteins that cause fatal neurodegenerative diseases. Because astrocytic gliosis marked by the deposition of fibrils composed of GFAP is a prominent feature of prion disease, we asked whether GFAP might be used as a surrogate marker for prions. To interrogate this posit, we inoculated prions into transgenic (Tg) mice expressing luciferase (luc) under the GFAP gene (Gfap) promoter, denoted Tg(Gfap-luc) mice. Weekly noninvasive, bioluminescence imaging (BLI) detected an increase in light emitted from the brains of Tg(Gfap-luc) mice at approximately 55 d after inoculation and approximately 62 d before neurologic deficits appeared. To determine whether BLI could be used as a proxy bioassay for prion infectivity, we performed endpoint titrations of prions in Tg(Gfap-luc) mice. BLI bioassays were as or more sensitive than those determined by the onset of neurological dysfunction, and were completed in approximately half the time. Our studies argue that BLI is likely to be a suitable surrogate for measuring prion infectivity, and might be useful in the study of Tg mouse models for other neurodegenerative illnesses.
Collapse
|
27
|
Pradines E, Loubet D, Mouillet-Richard S, Manivet P, Launay JM, Kellermann O, Schneider B. Cellular prion protein coupling to TACE-dependent TNF-α shedding controls neurotransmitter catabolism in neuronal cells. J Neurochem 2009; 110:912-23. [DOI: 10.1111/j.1471-4159.2009.06176.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
28
|
Jonakait GM, Ni L. Prostaglandins compromise basal forebrain cholinergic neuron differentiation and survival: action at EP1/3 receptors results in AIF-induced death. Brain Res 2009; 1285:30-41. [PMID: 19555672 DOI: 10.1016/j.brainres.2009.06.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 06/11/2009] [Accepted: 06/12/2009] [Indexed: 01/26/2023]
Abstract
Activated microglia produce a factor or cocktail of factors that promotes cholinergic neuronal differentiation of undifferentiated precursors in the embryonic basal forebrain (BF) in vitro. To determine whether microglial prostaglandins mediate this action, microglia were stimulated in the presence of the cyclooxygenase inhibitor ibuprofen, and microglial conditioned medium (CM) was used to culture rat BF precursors at embryonic day 15. Choline acetyltransferase (ChAT) activity served as a measure of cholinergic differentiation. While inhibition of prostaglandin biosynthesis did not affect the ability of microglial CM to promote ChAT activity, treatment of microglia with prostaglandin E2 (PGE2) inhibited it. Agonists of E prostanoid receptors EP2 (butaprost) and EP1/3 (sulprostone) mimicked PGE2, while misoprostol (E1-4) actually enhanced the action of CM. PGE2 added directly to BF cultures together with microglial CM also inhibited ChAT activity. While BF cultures expressed all four prostanoid receptors, direct addition of sulprostone but not butaprost mimicked PGE2, suggesting that PGE2 engaged EP1/3 receptors in the BF. Neither PKA inhibition by H89 nor cAMP induction by forskolin or dibutyrl-cAMP altered the action of sulprostone. Sulprostone severely compromised ChAT activity, dendrite number, axonal length and axonal branching, but caspase inhibition did not restore these. However, sulprostone resulted in increased staining intensity and nuclear translocation of apoptosis-inducing factor (AIF) suggesting caspase-independent cell death. We have found that PGE2 action at microglial EP2 receptors inhibits the microglial production of the cholinergic differentiating cocktail, while action at neuronal EP3 receptors has a deleterious effect on cholinergic neurons causing neurite retraction and cell death.
Collapse
Affiliation(s)
- G Miller Jonakait
- Federated Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | | |
Collapse
|
29
|
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: 2.2] [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
|
30
|
Kaneko M, Sugiyama N, Sasayama D, Yamaoka K, Miyakawa T, Arima K, Tsuchiya K, Hasegawa K, Washizuka S, Hanihara T, Amano N, Yagishita S. Prion disease causes less severe lesions in human hippocampus than other parts of brain. Psychiatry Clin Neurosci 2008; 62:264-70. [PMID: 18588585 DOI: 10.1111/j.1440-1819.2008.01792.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
AIM The hippocampus can be very sensitive to damage in the scrapie-infected mouse, a well-established animal model of prion diseases. Terminally ill scrapie-infected animals exhibit nearly complete loss of cornu ammonis (CA) 1 pyramidal neurons, but few studies have focused on the neuropathological lesions of the human hippocampus in autopsied brain tissue; in particular, few findings on differences in severity of pathology between the hippocampal and parahippocampal formations have been obtained. The aim of the present paper is to evaluate the human hippocampus of prion disease through neuropathological examination. METHODS A systemic, detailed neuropathological study throughout the subdivisions of the hippocampus was carried out in 23 autopsied cases of prion diseases. Prion protein immunohistochemistry was performed in serial brain sections to determine the topography of prion deposits. RESULTS Compared to lesions in other brain regions, hippocampal lesions were mild, despite numerous prion deposits. The distribution of prion deposits did not appear to be correlated with neuropathological changes. The present findings differed from the hippocampal pathology observed in scrapie-infected mice. In addition, differences in neuropathological severity were observed within the hippocampal formation. CONCLUSION The human hippocampus may be protected from the neurotoxic effects of prion deposits.
Collapse
Affiliation(s)
- Minoru Kaneko
- Department of Neuropsychiatry, Shinshu University School of Medicine, Nagano, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
LaCasse RA, Striebel JF, Favara C, Kercher L, Chesebro B. Role of Erk1/2 activation in prion disease pathogenesis: absence of CCR1 leads to increased Erk1/2 activation and accelerated disease progression. J Neuroimmunol 2008; 196:16-26. [PMID: 18396336 DOI: 10.1016/j.jneuroim.2008.02.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Revised: 01/16/2008] [Accepted: 02/11/2008] [Indexed: 12/14/2022]
Abstract
Prion diseases are neurodegenerative infections with gliosis and vacuolation. The mechanisms of degeneration remain unclear, but chemokines may be important. In current experiments CCR1 knock-out (KO) mice succumbed more rapidly to scrapie infection than WT controls. Infected KO mice had upregulation of CCL3, a CCR1 ligand, and CCR5, a receptor with specificity for CCL3. Both infected KO and WT mice had upregulation of CCR5-mediated signaling involving activation of Erk1/2 in astrocytes; however, activation was earlier in KO mice suggesting a role in pathogenesis. In both mouse strains activation of the Erk1/2 pathway may lead to astrocyte dysfunction resulting in neurodegeneration.
Collapse
Affiliation(s)
- Rachel A LaCasse
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, United States
| | | | | | | | | |
Collapse
|
32
|
Solito E, McArthur S, Christian H, Gavins F, Buckingham JC, Gillies GE. Annexin A1 in the brain--undiscovered roles? Trends Pharmacol Sci 2008; 29:135-42. [PMID: 18262660 DOI: 10.1016/j.tips.2007.12.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Revised: 12/05/2007] [Accepted: 12/06/2007] [Indexed: 11/16/2022]
Abstract
Annexin A1 (ANXA1) is an endogenous protein known to have potent anti-inflammatory properties in the peripheral system. It has also been detected in the brain, but its function there is still ambiguous. In this review, we have, for the first time, collated the evidence currently available on the function of ANXA1 in the brain and have proposed several possible mechanisms by which it exerts a neuroprotective or anti-neuroinflammatory function. We suggest that ANXA1, its small peptide mimetics and its receptors might be exciting new therapeutic targets in the management of a wide range of neuroinflammatory diseases, including stroke and neurodegenerative conditions.
Collapse
Affiliation(s)
- Egle Solito
- Imperial College London, Commonwealth Building, Hammersmith Hospital, DuCane Road, London, UK.
| | | | | | | | | | | |
Collapse
|
33
|
Motoyoshi A, Nakajima H, Takano K, Moriyama M, Kannan Y, Nakamura Y. Effects of Amphotericin B on the expression of neurotoxic and neurotrophic factors in cultured microglia. Neurochem Int 2008; 52:1290-6. [PMID: 18328601 DOI: 10.1016/j.neuint.2008.01.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 01/18/2008] [Accepted: 01/24/2008] [Indexed: 12/01/2022]
Abstract
Amphotericin B (AmB) is a polyene antibiotic and reported to have therapeutic effects on prion diseases, in which the microglial activation has been suggested to play important roles by proliferating and producing various factors such as nitric oxide, proinflammatory cytokines, and so on. However, the therapeutic mechanism of AmB on prion diseases remains elusive. In the present study, we investigated the effects of AmB on cellular functions of rat primary cultured microglia. We found that AmB, similarly as lipopolysaccharide (LPS), could activate microglia to produce nitric oxide via inducible nitric oxide synthase. Both AmB and LPS also induced mRNA expressions of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in microglia. AmB also changed the expression levels of neurotrophic factors mRNAs. AmB and LPS significantly down-regulated the level of ciliary neurotrophic factor mRNA. However, AmB, but not LPS, significantly up-regulated the level of glial cell-line derived neurotrophic factor mRNA in microglia. In addition, brain-derived neurotrophic factor mRNA expression level was tending upward by treatment with AmB, but not with LPS. Taken together, these results suggest that AmB regulates the microglial activation in different manner from LPS and that microglia may participate in the therapeutic effects of AmB on prion diseases by controlling the expression and production of such mediators.
Collapse
Affiliation(s)
- Akiko Motoyoshi
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Prefecture University 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Bedecs K. Cell culture models to unravel prion protein function and aberrancies in prion diseases. Methods Mol Biol 2008; 459:1-20. [PMID: 18576144 DOI: 10.1007/978-1-59745-234-2_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
From an early stage of prion research, tissue cultures that could support and propagate the scrapie agent were sought after. The earliest attempts were explants from brains of infected mice, and their growth and morphological characteristics were compared with those from uninfected mice. Using the explant technique, several investigators reported increased cell growth in cultures established from scrapie-sick brain compared with cultures from normal mice. These are odd findings in the light of the massive neuronal cell death known to occur in scrapie-infected brains; however, the cell types responsible for the increased cell growth in the scrapie-explants most probably were not neuronal. The first successful cell culture established in this way, in which the scrapie agent was serially and continuously passaged beyond the initial explant, was in the scrapie mouse brain culture, which is still used today. This chapter describes the generation and use of chronically prion-infected cell lines as cell culture models of prion diseases. These cell lines have been crucial for the current understanding of the cell biology of both the normal (PrP(C)) and the pathogenic isoform (PrP(Sc)) of the prion protein. They also have been useful in the development of antiprion drugs, prospectively used for therapy of prion diseases, and they offer an alternative approach for transmission/infectivity assays normally performed by mouse bioassay. Cell culture models also have been used to study prion-induced cytopathological changes, which could explain the typical spongiform neurodegeneration in prion diseases.
Collapse
Affiliation(s)
- Katarina Bedecs
- Department of Biochemistry and Biophysics, Stockholm University, Sweden
| |
Collapse
|
35
|
Jinno S, Fleischer F, Eckel S, Schmidt V, Kosaka T. Spatial arrangement of microglia in the mouse hippocampus: a stereological study in comparison with astrocytes. Glia 2007; 55:1334-47. [PMID: 17647290 DOI: 10.1002/glia.20552] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Microglia are classically considered to be immune cells in the brain, but have now been proven to be involved in neuronal activity as well. Here we stereologically analyzed the spatial arrangement of microglia in the mouse hippocampus. First, we estimated the numerical densities (NDs) of microglia identified by ionized calcium-binding adaptor molecule 1 (Iba1). Despite that microglia appeared to be evenly distributed throughout the hippocampal area, the NDs demonstrated significant dorsoventral, interregional, and interlaminar differences. Briefly, the NDs in the ventral hippocampus were significantly lower in the CA3 region than in the CA1 region and dentate gyrus, although no interregional differences were detectable in the dorsal hippocampus. Both in the CA1 and CA3 regions, the NDs were significantly higher in the stratum lacunosum-moleculare than in the remaining layers. Next, we investigated the spatial patterns of distribution of Iba1-labeled microglia and S100beta-labeled astrocytes. So far as we examined, the somato-somatic contacts were not seen among microglia or among astrocytes, whereas the close apposition between microglia and astrocytes were occasionally detected. The 3D point process analysis showed that the spatial distribution of microglia was significantly repulsive. Because the statistical territory of single microglia was larger than that estimated from process tracing, they are not likely to touch each other with their processes. Astrocytes were distributed slightly repulsively with overlapping areas. The 3D point process analysis also revealed a significant spatial attraction between microglia and astrocytes. The present findings provide a novel anatomical basis for glial research.
Collapse
Affiliation(s)
- Shozo Jinno
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan.
| | | | | | | | | |
Collapse
|
36
|
Kim JI, Jin JK, Choi EK, Spinner D, Rubenstein R, Carp RI, Kim YS. Increased expression and localization of cyclooxygenase-2 in astrocytes of scrapie-infected mice. J Neuroimmunol 2007; 187:74-82. [PMID: 17524497 DOI: 10.1016/j.jneuroim.2007.04.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Accepted: 04/10/2007] [Indexed: 02/02/2023]
Abstract
A number of aspects of the pathogenesis of scrapie, the archetype disease of the transmissible spongiform encephalopathies (prion disorders), remain to be elucidated. There is increasing evidence that there are cerebral based inflammatory processes that may contribute to the pathogenesis and to the progression of a number of neurodegenerative disorders, including prion diseases. In peripheral tissues, a key element that controls the generation of proinflammatory mediators is the highly inducible protein cyclooxygenase-2 (COX-2). In this study, in order to examine the possible association of COX-2 with the pathogenesis of scrapie, we analyzed the expression level and the cellular localization of COX-2 in the brains of control and scrapie-infected mice. The COX-2 mRNA and protein levels were increased significantly compared to the control group of mice. By immunohistological analysis, intense immunoreactivity of COX-2 was localized primarily in reactive astrocytes, with virtually no staining in sections from control mice. The staining for COX-2 was co-localized with the pathological form of the prion protein (PrP(Sc)) and with nuclear factor-kappa B (NF-kappaB). These results suggest that the upregulation of COX-2 expression in astrocytes may be related to the accumulation of PrP(Sc), and that COX-2 may then lead to the progression of scrapie, possibly by propagation of a cerebral inflammatory response.
Collapse
Affiliation(s)
- Jae-Il Kim
- New York State Institute for Basic Research, Staten Island, New York, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Jin JK, Na YJ, Song JH, Joo HG, Kim S, Kim JI, Choi EK, Carp RI, Kim YS, Shin T. Galectin-3 expression is correlated with abnormal prion protein accumulation in murine scrapie. Neurosci Lett 2007; 420:138-43. [PMID: 17531384 DOI: 10.1016/j.neulet.2007.04.069] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 04/21/2007] [Accepted: 04/23/2007] [Indexed: 11/23/2022]
Abstract
To investigate the involvement of galectin-3 in the process of neurodegeneration in prion diseases, the expression and cellular localization of galectin-3 in the brain were studied in scrapie, a mouse model of prion disease. Reverse transcription-polymerase chain reaction (RT-PCR) and Western blot analyses showed that the expression of galectin-3 protein and mRNA was induced in scrapie-affected brains, particularly at the time when the abnormal prion protein PrP(Sc) began to accumulate in the brains. Immunohistochemically, immunostaining for galectin-3 was found mainly in B4-isolectin-positive cells (presumably activated microglia/macrophages), but not in astrocytes. Galectin-3 immunoreactivity was localized mainly in areas of PrP(Sc) accumulation and neuronal death in scrapie-infected brains. These findings suggest that the expression of galectin-3 by activated microglia/macrophages in prion disease correlates with abnormal prion protein accumulation.
Collapse
Affiliation(s)
- Jae-Kwang Jin
- Ilsong Institute of Life Science, Hallym University Medical Center, Anyang, Kyonggi-do 431-060, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Marcos-Carcavilla A, Calvo JH, González C, Moazami-Goudarzi K, Laurent P, Bertaud M, Hayes H, Beattie AE, Serrano C, Lyahyai J, Martín-Burriel I, Alves E, Zaragoza P, Badiola JJ, Serrano M. IL-1 family members as candidate genes modulating scrapie susceptibility in sheep: localization, partial characterization, and expression. Mamm Genome 2007; 18:53-63. [PMID: 17242860 DOI: 10.1007/s00335-006-0095-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Accepted: 10/30/2006] [Indexed: 01/08/2023]
Abstract
Scrapie (SC) is a transmissible spongiform encephalopathy (TSE) in sheep and goats. Susceptibility to this neurodegenerative disease is controlled mainly by point mutations at the PRNP locus. Other genes, apart from PRNP, have been reported to modulate resistance/susceptibility to SC. On the basis of several studies on Alzheimer's disease and different TSE models, and of requirement for correct homeostasis of cytokines in brain, IL1B and IL1RN were chosen as putative positional and functional candidate genes that might be involved in the polygenic variance mentioned above. In the present work, ovine IL1B and IL1RN genes were partially isolated and characterized, including promoter and other regulatory regions. In addition, several sequence polymorphisms were identified. Furthermore, their cytogenetic positions on sheep chromosomes were determined by FISH and confirmed by linkage analysis, localizing both genes in OAR3p22, a region previously described as carrying a QTL for SC incubation period in sheep. Finally, expression analyses were carried out in eight naturally SC-infected and five uninfected sheep with the same genotype for PRNP (ARQ/ARQ). This comparison was performed using real-time RT-PCR in samples of spleen and cerebellum. Results showed differences in the expression of both cytokines in cerebellum (p < 0.05) but not in spleen (p > 0.05).
Collapse
Affiliation(s)
- Ane Marcos-Carcavilla
- Departamento de Mejora Genética Animal, INIA, Ctra La Coruña Km 7.5, 28040, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Pasquali P, Nonno R, Mandara MT, Di Bari MA, Ricci G, Petrucci P, Capuccini S, Cartoni C, Macrì A, Agrimi U. Intracerebral administration of interleukin-12 (IL-12) and IL-18 modifies the course of mouse scrapie. BMC Vet Res 2006; 2:37. [PMID: 17192191 PMCID: PMC1769363 DOI: 10.1186/1746-6148-2-37] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Accepted: 12/27/2006] [Indexed: 11/10/2022] Open
Abstract
Background Prion diseases are characterised by a neurodegenerative pattern in which the function of immune system remains still elusive. In the present study, we evaluate if an exogenous treatment with Interleukin-12 (IL-12) and IL-18, able to activate microglia, is able to affect scrapie pathogenesis. Results Cytokines injected intracranially, induced a strong inflammatory response characterised by TNF-α production and microglia activation. Two groups of mice were injected intracerebrally with high dose of ME7 strain of scrapie containing IL-12 and IL-18 or sterile saline. Cytokines-treated mice showed a more pronounced accumulation of PrPSc in brain tissues at 90 days post-inoculation and a shorter mean survival times than untreated mice. Conclusion We can conclude that intracerebral administration of IL-12 and IL-18 can modulate scrapie pathogenesis possibly through a microglia-mediated pattern.
Collapse
Affiliation(s)
- Paolo Pasquali
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Romolo Nonno
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Maria Teresa Mandara
- Department of Biopathological Veterinary Science, Veterinary Medicine School, Università degli Studi di Perugia, Perugia, Italy
| | - Michele Angelo Di Bari
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Giovanni Ricci
- Department of Biopathological Veterinary Science, Veterinary Medicine School, Università degli Studi di Perugia, Perugia, Italy
| | - Paola Petrucci
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Silvia Capuccini
- Department of Biopathological Veterinary Science, Veterinary Medicine School, Università degli Studi di Perugia, Perugia, Italy
| | - Claudia Cartoni
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Agostino Macrì
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Umberto Agrimi
- Department of Food Safety and Animal Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
40
|
Schneider B, Pietri M, Mouillet-Richard S, Ermonval M, Mutel V, Launay JM, Kellermann O. Control of Bioamine Metabolism by 5-HT2Band α1DAutoreceptors through Reactive Oxygen Species and Tumor Necrosis Factor-α Signaling in Neuronal Cells. Ann N Y Acad Sci 2006; 1091:123-41. [PMID: 17341609 DOI: 10.1196/annals.1378.061] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Homeostasis of the central nervous system relies on the proper integration of cell-signaling pathways recruited by a variety of neuronal and non-neuronal factors, with the aim of tightly controlling neurotransmitter metabolism, storage, and transport. We took advantage of the 1C11 neuroectodermal cell line, endowed with the capacity to selectively differentiate into serotonergic (1C11(5-HT)) or noradrenergic (1C11(NE)) neurons, to identify functional targets of serotonin (5-hydroxytryptamine [5-HT]) and norepinephrine (NE) autoreceptors possibly involved in the control of neuronal functions. We demonstrate that 5-HT(2B) and adreno alpha(1D) receptors are coupled to reactive oxygen species (ROS) production through NADPH oxidase activation in 1C11(5-HT) and 1C11(NE) neuronal cells, respectively. In the signaling cascade linking 5-HT(2B) receptors to NADPH oxidase, phospholipase A2-mediated arachidonic acid production is required for ROS synthesis. ROS, in turn, act as second message signals and control the activation of TACE (TNF-alpha converting enzyme), a member of a disintegrin and metalloproteinase family. 5-HT(2B) and alpha(1D) receptor stimulation triggers TACE-dependent TNF-alpha shedding in the surrounding milieu of 1C11(5-HT) and 1C11(NE) cells. In these cells, shed TNF-alpha triggers degradation of 5-HT and NE into 5-HIAA and MHPG, respectively. Finally, we observe that 5-HT(2B) and alpha(1D) receptor couplings to the NADPH oxidase-TACE cascade are strictly restricted to 1C11-derived progenies that have implemented a complete neuronal phenotype. Altogether, our data indicate that couplings of 5-HT(2B) and alpha(1D) autoreceptors to ROS and TNF-alpha signaling control neurotransmitter metabolism in 1C11-derived neuronal cells. Eventually, we might explain the origin of oxidative stress and high level of TNF-alpha in neurodegenerative diseases as a consequence of deviation of normal signaling pathways coupled to neurotransmitters.
Collapse
Affiliation(s)
- Benoit Schneider
- Institut André Lwoff-Institut Pasteur, CNRS FRE 2937, Laboratoire Différenciation Cellulaire et Prions, 7 rue Guy Môquet, 94801 Villejuif Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
41
|
Iwamaru Y, Takenouchi T, Ogihara K, Hoshino M, Takata M, Imamura M, Tagawa Y, Hayashi-Kato H, Ushiki-Kaku Y, Shimizu Y, Okada H, Shinagawa M, Kitani H, Yokoyama T. Microglial cell line established from prion protein-overexpressing mice is susceptible to various murine prion strains. J Virol 2006; 81:1524-7. [PMID: 17121794 PMCID: PMC1797525 DOI: 10.1128/jvi.01379-06] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Several lines of evidence suggest that microglia have important roles in the pathogenesis of prion diseases. Here, we establish a novel microglial cell line (MG20) from neonatal tga20 mice that overexpress murine prion protein. After exposure to Chandler scrapie, we observed the replication and accumulation of disease-associated forms of the prion protein in MG20 cells up to the 15th passage. Furthermore, MG20 cells were susceptible to ME7, Obihiro scrapie, and bovine spongiform encephalopathy agents. Thus, MG20 cell lines persistently infected with various murine prion strains provide a useful model for the study of the pathogenesis of prion diseases.
Collapse
Affiliation(s)
- Yoshifumi Iwamaru
- Research Center for Prion Diseases, National Institute of Animal Health, Kannondai 3-1-5, Tsukuba, Ibaraki 305-8602, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Eikelenboom P, Veerhuis R, Scheper W, Rozemuller AJM, van Gool WA, Hoozemans JJM. The significance of neuroinflammation in understanding Alzheimer's disease. J Neural Transm (Vienna) 2006; 113:1685-95. [PMID: 17036175 DOI: 10.1007/s00702-006-0575-6] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2006] [Accepted: 09/05/2006] [Indexed: 01/26/2023]
Abstract
The interest of scientists in the involvement of inflammation-related mechanisms in the pathogenesis of Alzheimer's disease (AD) goes back to the work of one of the pioneers of the study of this disease. About hundred years ago Oskar Fischer stated that the crucial step in the plaque formation is the extracellular deposition of a foreign substance that provokes an inflammatory reaction followed by a regenerative response of the surrounding nerve fibers. Eighty years later immunohistochemical studies revealed that amyloid plaques are indeed co-localized with a broad variety of inflammation-related proteins (complement factors, acute-phase proteins, pro-inflammatory cytokines) and clusters of activated microglia. These findings have led to the view that the amyloid plaque is the nidus of a non-immune mediated chronic inflammatory response locally induced by fibrillar A beta deposits. Recent neuropathological studies show a close relationship between fibrillar A beta deposits, inflammation and neuroregeneration in relatively early stages of AD pathology preceding late AD stages characterized by extensive tau-related neurofibrillary changes. In the present work we will review the role of inflammation in the early stage of AD pathology and particularly the role of inflammation in A beta metabolism and deposition. We also discuss the possibilities of inflammation-based therapeutic strategies in AD.
Collapse
Affiliation(s)
- P Eikelenboom
- Department of Neurology, Academic Medical Center, University of Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
43
|
Jin JK, Na YJ, Moon C, Kim H, Ahn M, Kim YS, Shin T. Increased expression of osteopontin in the brain with scrapie infection. Brain Res 2006; 1072:227-33. [PMID: 16412998 DOI: 10.1016/j.brainres.2005.12.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Revised: 11/24/2005] [Accepted: 12/05/2005] [Indexed: 11/24/2022]
Abstract
The expression of osteopontin (OPN) was studied in the brains of mice with scrapie. Reverse transcriptase polymerase chain reaction (RT-PCR) and Western blot analysis showed that the expression of OPN protein and mRNA was increased significantly in the scrapie-infected brains compared to the controls. The increased expression of OPN protein was largely matched with the PrP(Sc) accumulation. Immunohistochemically, OPN was intensely immunostained in neurons of the midbrain at the time of scrapie infection initiation. Particularly, OPN immunostaining was noted in the reactive astrocytes and some microglia in the scrapie brains, while those cells were devoid of OPN immunoreactivity in control brains. Overall, these findings suggest that some neurons affected by PrP(Sc) at an early stage of scrapie transiently express OPN but subsequently succumb to cell death at a later stage of scrapie; astroglial cells after scrapie infection are activated to express OPN; and increased OPN expression in these cells may play an important role in the pathology of scrapie. The precise role of OPN in scrapie needs further study.
Collapse
Affiliation(s)
- Jae-Kwang Jin
- Ilsong Institute of Life Science, Hallym University, Ilsong Building, 1605-4 Kwanyang-dong, Dongan-gu, Anyang, Kyonggi-do 431-060, South Korea
| | | | | | | | | | | | | |
Collapse
|
44
|
Bate C, Kempster S, Williams A. Prostaglandin D2 mediates neuronal damage by amyloid-β or prions which activates microglial cells. Neuropharmacology 2006; 50:229-37. [PMID: 16289250 DOI: 10.1016/j.neuropharm.2005.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Revised: 08/23/2005] [Accepted: 09/26/2005] [Indexed: 11/22/2022]
Abstract
Microglial cells killed neurons damaged following incubation with sub-lethal concentrations of peptides derived from either the human prion protein (HuPrP82-146) or amyloid-beta1-42 (a peptide found in Alzheimer's disease). HuPrP82-146 or amyloid-beta1-42 induced phenotypic changes in neurons that caused them to bind a CD14-IgG chimera. In co-cultures microglial cells produced interleukin (IL)-6 in response to HuPrP82-146 or amyloid-beta1-42 damaged neurons. The binding of the CD14-IgG chimera to HuPrP82-146 or amyloid-beta1-42 damaged neurons was reduced by pre-treatment with cyclo-oxygenase (COX)-1 inhibitors and in co-cultures, COX-1 inhibitors significantly increased neuronal survival. Studies with individual prostaglandins demonstrated that the addition of prostaglandin D2, or prostaglandin E2, but not other prostaglandins (F2alpha, H2, I2 or 15-dJ2), mimicked the effects of amyloid-beta1-42 on neurons. Thus, prostaglandin D2 or E2 damaged neurons bound the CD14-IgG chimera, and in co-cultures prostaglandin D2 damaged neurons activated microglial cells. These effects were mediated via the DP prostanoid receptor; DP receptor agonists BW245C or SQ27986 induced neuronal damage, while the DP receptor antagonist BWA868C was neuroprotective in co-cultures. These results indicate that prostaglandin D2, produced following activation of COX-1 by sub-lethal concentrations of HuPrP82-146 or amyloid-beta1-42, causes phenotypic changes in neurons that activates microglial cells and leads to neuronal loss.
Collapse
Affiliation(s)
- Clive Bate
- Department of Pathology and Infectious Diseases, Royal Veterinary College, Hawkshead Lane, North Mymms, Herts, AL9 7TA, UK.
| | | | | |
Collapse
|
45
|
Garção P, Oliveira CR, Agostinho P. Comparative study of microglia activation induced by amyloid-beta and prion peptides: Role in neurodegeneration. J Neurosci Res 2006; 84:182-93. [PMID: 16634056 DOI: 10.1002/jnr.20870] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The inflammatory responses in Alzheimer's disease (AD) and prion-related encephalopathies (PRE) are dominated by microglia activation. Several studies have reported that the amyloid-beta (Abeta) peptides, which are associated with AD, and the pathogenic isoform of prion protein (PrPSc) have a crucial role in neuronal death and gliosis that occur in both of these disorders. In this study, we investigate whether Abeta and PrPSc cause microglia activation per se and whether these amyloidogenic peptides differentially affect these immunoeffector cells. In addition, we also determined whether substances released by Abeta- and PrP-activated microglia induce neuronal death. Cultures of rat brain microglia cells were treated with the synthetic peptides Abeta1-40, Abeta1-42 and PrP106-126 for different time periods. The lipopolysaccharide was used as a positive control of microglia activation. Our results show that Abeta1-40 and PrP106-126 caused similar morphological changes in microglia and increased the production of nitric oxide and hydroperoxides. An increase on inducible nitric oxide synthase expression was also observed in microglia treated with Abeta1-40 or PrP106. However, these peptides affected in a different manner the secretion of interleukin-1beta (IL-1beta) and interleukin-6 (IL-6) secretion. In cocultures of microglia-neurons, it was observed that microglia treated with Abeta1-40 or PrP106-126 induced a comparable extent of neuronal death. The neutralizing antibody for IL-6 significantly reduced the neuronal death induced by Abeta- or PrP-activated microglia. Taken together, the data indicate that Abeta and PrP peptides caused microglia activation and differentially affected cytokine secretion. The IL-6 released by reactive microglia caused neuronal injury.
Collapse
Affiliation(s)
- Pedro Garção
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | | |
Collapse
|
46
|
Hoozemans JJM, Veerhuis R, Rozemuller JM, Eikelenboom P. Neuroinflammation and regeneration in the early stages of Alzheimer's disease pathology. Int J Dev Neurosci 2005; 24:157-65. [PMID: 16384684 DOI: 10.1016/j.ijdevneu.2005.11.001] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Accepted: 11/10/2005] [Indexed: 10/25/2022] Open
Abstract
The initial stages of Alzheimer's disease pathology in the neocortex show upregulation of cell cycle proteins, adhesion and inflammation related factors, indicating the early involvement of inflammatory and regenerating pathways in Alzheimer's disease pathogenesis. These brain changes precede the neurofibrillary pathology and the extensive process of neurodestruction and (astro)gliosis. Amyloid beta deposition, inflammation and regenerative mechanisms are also early pathogenic events in transgenic mouse models harbouring the pathological Alzheimer's disease mutations, while neurodegenerative characteristics are not seen in these models. This review will discuss the relationship between neuroinflammation and neuroregeneration in the early stages of Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
- J J M Hoozemans
- Department of Neuropathology, Academic Medical Center, University of Amsterdam, P.O. Box 22700, 110DE Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
47
|
Puoti G, Giaccone G, Mangieri M, Limido L, Fociani P, Zerbi P, Suardi S, Rossi G, Iussich S, Capobianco R, Di Fede G, Marcon G, Cotrufo R, Filippini G, Bugiani O, Tagliavini F. Sporadic Creutzfeldt-Jakob disease: the extent of microglia activation is dependent on the biochemical type of PrPSc. J Neuropathol Exp Neurol 2005; 64:902-9. [PMID: 16215462 DOI: 10.1097/01.jnen.0000183346.19447.55] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
In prion-related encephalopathies, microglial activation occurs early and is dependent on accumulation of disease-specific forms of the prion protein (PrPSc) and may play a role in nerve cell death. Previously, we found that different types of PrPSc (i.e. type 1 and type 2) coexisted in approximately 25% of patients with sporadic Creutzfeldt-Jakob disease (CJD); and a close relationship was detected between PrPSc type, the pattern of PrP immunoreactivity, and extent of spongiform degeneration. To investigate whether microglial reaction is related to the biochemical type and deposition pattern of PrPSc, we carried out a neuropathologic and biochemical study on 26 patients with sporadic CJD, including all possible genotypes at codon 129 of the prion protein gene. By quantitative analysis, we demonstrated that strong microglial activation was associated with type 1 PrPSc and diffuse PrP immunoreactivity, whereas type 2 PrPSc and focal PrP deposits were accompanied by mild microglia reaction. These findings support the view that the phenotypic heterogeneity of sporadic CJD is largely determined by the physicochemical properties of distinct PrPSc conformers.
Collapse
Affiliation(s)
- Gianfranco Puoti
- Istituto Nazionale Neurologico Carlo Besta, Milano, and Dipartimento di Scienze Neurologiche, Seconda Università di Napoli, Napoli, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Veerhuis R, Boshuizen RS, Morbin M, Mazzoleni G, Hoozemans JJM, Langedijk JPM, Tagliavini F, Langeveld JPM, Eikelenboom P. Activation of human microglia by fibrillar prion protein-related peptides is enhanced by amyloid-associated factors SAP and C1q. Neurobiol Dis 2005; 19:273-82. [PMID: 15837583 DOI: 10.1016/j.nbd.2005.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2004] [Revised: 12/06/2004] [Accepted: 01/04/2005] [Indexed: 11/19/2022] Open
Abstract
Complement activation products C1q and C3d, serum amyloid P component (SAP) and activated glial cells accumulate in amyloid deposits of conformationally changed prion protein (PrP(Sc)) in Creutzfeldt-Jakob disease, Gerstmann-Sträussler-Scheinker disease and scrapie-infected mouse brain. Biological properties, including the potential to activate microglia, relate to prion (PrP) peptide fibrillogenic abilities. We investigated if SAP and C1q influence the fibrillogenic properties of human and mouse PrP peptide and concomitantly their stimulatory effects on human microglia in vitro. PrP-peptide induced microglial IL-6 and TNF-alpha release significantly increased in the presence of SAP and C1q. Also, SAP and C1q enhanced PrP-peptide fibril formation as revealed by electron microscopy and thioflavin S-based quantitative assays. This suggests that SAP and C1q contribute to fibrillar state-dependent cellular effects of PrP. Combined, ultrastructural and thioflavin assays, together with microglial cytokine release measurements, provide a test system to screen potential, fibrillarity impeding therapeutics for prion disease.
Collapse
Affiliation(s)
- Robert Veerhuis
- Institute for Clinical and Experimental Neurosciences-VU (ICEN-VU), Department of Psychiatry, Vrije Universiteit University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Pietri M, Schneider B, Mouillet-Richard S, Ermonval M, Mutel V, Launay JM, Kellermann O. Reactive oxygen species‐dependent TNF‐α converting enzyme activation through stimulation of 5‐HT2Band α1Dautoreceptors in neuronal cells. FASEB J 2005; 19:1078-87. [PMID: 15985531 DOI: 10.1096/fj.04-3631com] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A major determinant of neuronal homeostasis is the proper integration of cell signaling pathways recruited by a variety of neuronal and non-neuronal factors. By taking advantage of a neuroectodermal cell line (1C11) endowed with the capacity to differentiate into serotonergic (1C115-HT) or noradrenergic (1C11NE) neurons, we identified serotonin (5-hydroxytryptamine, 5-HT)- and norepinephrine (NE)-dependent signaling cascades possibly involved in neuronal functions. First, we establish that 5-HT2B receptors and 1D adrenoceptors are functionally coupled to reactive oxygen species (ROS) synthesis through NADPH oxidase activation in 1C115-HT and 1C11NE cells. This observation constitutes the prime evidence that bioaminergic autoreceptors take part in the control of the cellular redox equilibrium in a neuronal context. Second, our data identify TACE (TNF- Converting Enzyme), a member of a disintegrin and metalloproteinase (ADAM) family, as a downstream target of the 5-HT2B and 1D receptor-NADPH oxidase signaling pathways. Upon 5-HT2B or 1D receptor stimulation, ROS fully govern TNF- shedding in the surrounding milieu of 1C115-HT or 1C11NE cells. Third, 5-HT2B and 1Dreceptor couplings to the NADPH oxidase-TACE cascade are strictly restricted to 1C11-derived progenies that have implemented a complete serotonergic or noradrenergic phenotype. Overall, these observations suggest that 5-HT2B and 1D autoreceptors may play a role in the maintenance of neuron- and neurotransmitter-associated functions. Eventually, our study may have implications regarding the origin of oxidative stress as well as up-regulated expression of proinflammatory cytokines in neurodegenerative disorders, which may relate to the deviation of normal signaling pathways.
Collapse
Affiliation(s)
- Mathéa Pietri
- Institut André Lwoff-Institut Pasteur, CNRS UPR 1983, Laboratoire de Différenciation Cellulaire et Prions, Villejuif Cedex, France
| | | | | | | | | | | | | |
Collapse
|
50
|
Unterberger U, Voigtländer T, Budka H. Pathogenesis of prion diseases. Acta Neuropathol 2005; 109:32-48. [PMID: 15645262 DOI: 10.1007/s00401-004-0953-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2004] [Accepted: 10/18/2004] [Indexed: 11/28/2022]
Abstract
Prion diseases are rare neurological disorders that may be of genetic or infectious origin, but most frequently occur sporadically in humans. Their outcome is invariably fatal. As the responsible pathogen, prions have been implicated. Prions are considered to be infectious particles that represent mainly, if not solely, an abnormal, protease-resistant isoform of a cellular protein, the prion protein or PrP(C). As in other neurodegenerative diseases, aggregates of misfolded protein conformers are deposited in the CNS of affected individuals. Pathogenesis of prion diseases comprises mainly two equally important, albeit essentially distinct, topics: first, the mode, spread, and amplification of infectivity in acquired disease, designated as peripheral pathogenesis. In this field, significant advances have implicated an essential role of lymphoid tissues for peripheral prion replication, before a likely neural spread to the CNS. The second is the central pathogenesis, dealing, in addition to spread and replication of prions within the CNS, with the mechanisms of nerve cell damage and death. Although important roles for microglial neurotoxicity, oxidative stress, and complement activation have been identified, we are far from complete understanding, and therapeutic applications in prion diseases still need to be developed.
Collapse
|