48
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Chronic Toxoplasma gondii infection enhances β-amyloid phagocytosis and clearance by recruited monocytes

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction

          Alzheimer’s disease (AD) is associated with the accumulation of β-amyloid (Aβ) as senile plaques in the brain, thus leading to neurodegeneration and cognitive impairment. Plaque formation depends not merely on the amount of generated Aβ peptides, but more importantly on their effective removal. Chronic infections with neurotropic pathogens, most prominently the parasite Toxoplasma ( T.) gondii, are frequent in the elderly, and it has been suggested that the resulting neuroinflammation may influence the course of AD. In the present study, we investigated how chronic T. gondii infection and resulting neuroinflammation affect plaque deposition and removal in a mouse model of AD.

          Results

          Chronic infection with T. gondii was associated with reduced Aβ and plaque load in 5xFAD mice. Upon infection, myeloid-derived CCR2 hi Ly6C hi monocytes, CCR2 + Ly6C int, and CCR2 + Ly6C low mononuclear cells were recruited to the brain of mice. Compared to microglia, these recruited mononuclear cells showed highly increased phagocytic capacity of Aβ ex vivo. The F4/80 + Ly6C low macrophages expressed high levels of Triggering Receptor Expressed on Myeloid cells 2 (TREM2), CD36, and Scavenger Receptor A1 (SCARA1), indicating phagocytic activity. Importantly, selective ablation of CCR2 + Ly6C hi monocytes resulted in an increased amount of Aβ in infected mice. Elevated insulin-degrading enzyme ( IDE), matrix metalloproteinase 9 ( MMP9), as well as immunoproteasome subunits β 1i/LMP2, β2i/MECL-1, and β5i/LMP7 mRNA levels in the infected brains indicated increased proteolytic Aβ degradation. Particularly, LMP7 was highly expressed by the recruited mononuclear cells in the brain, suggesting a novel mechanism of Aβ clearance.

          Conclusions

          Our results indicate that chronic Toxoplasma infection ameliorates β-amyloidosis in a murine model of AD by activation of the immune system, specifically by recruitment of Ly6C hi monocytes and by enhancement of phagocytosis and degradation of soluble Aβ. Our findings provide evidence for a modulatory role of inflammation-induced Aβ phagocytosis and degradation by newly recruited peripheral immune cells in the pathophysiology of AD.

          Electronic supplementary material

          The online version of this article (doi:10.1186/s40478-016-0293-8) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references84

          • Record: found
          • Abstract: found
          • Article: not found

          Inflammation and Alzheimer's disease.

          Inflammation clearly occurs in pathologically vulnerable regions of the Alzheimer's disease (AD) brain, and it does so with the full complexity of local peripheral inflammatory responses. In the periphery, degenerating tissue and the deposition of highly insoluble abnormal materials are classical stimulants of inflammation. Likewise, in the AD brain damaged neurons and neurites and highly insoluble amyloid beta peptide deposits and neurofibrillary tangles provide obvious stimuli for inflammation. Because these stimuli are discrete, microlocalized, and present from early preclinical to terminal stages of AD, local upregulation of complement, cytokines, acute phase reactants, and other inflammatory mediators is also discrete, microlocalized, and chronic. Cumulated over many years, direct and bystander damage from AD inflammatory mechanisms is likely to significantly exacerbate the very pathogenic processes that gave rise to it. Thus, animal models and clinical studies, although still in their infancy, strongly suggest that AD inflammation significantly contributes to AD pathogenesis. By better understanding AD inflammatory and immunoregulatory processes, it should be possible to develop anti-inflammatory approaches that may not cure AD but will likely help slow the progression or delay the onset of this devastating disorder.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool.

            In multiple sclerosis and the experimental autoimmune encephalitis (EAE) mouse model, two pools of morphologically indistinguishable phagocytic cells, microglia and inflammatory macrophages, accrue from proliferating resident precursors and recruitment of blood-borne progenitors, respectively. Whether these cell types are functionally equivalent is hotly debated, but is challenging to address experimentally. Using a combination of parabiosis and myeloablation to replace circulating progenitors without affecting CNS-resident microglia, we found a strong correlation between monocyte infiltration and progression to the paralytic stage of EAE. Inhibition of chemokine receptor-dependent recruitment of monocytes to the CNS blocked EAE progression, suggesting that these infiltrating cells are essential for pathogenesis. Finally, we found that, although microglia can enter the cell cycle and return to quiescence following remission, recruited monocytes vanish, and therefore do not ultimately contribute to the resident microglial pool. In conclusion, we identified two distinct subsets of myelomonocytic cells with distinct roles in neuroinflammation and disease progression.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Microglia and neurodegeneration: the role of systemic inflammation.

              It is well accepted that CNS inflammation has a role in the progression of chronic neurodegenerative disease, although the mechanisms through which this occurs are still unclear. The inflammatory response during most chronic neurodegenerative disease is dominated by the microglia and mechanisms by which these cells contribute to neuronal damage and degeneration are the subject of intense study. More recently it has emerged that systemic inflammation has a significant role to play in the progression of these diseases. Well-described adaptive pathways exist to transduce systemic inflammatory signals to the brain, but activation of these pathways appears to be deleterious to the brain if the acute insult is sufficiently robust, as in severe sepsis, or sufficiently prolonged, as in repeated stimulation with robust doses of inflammogens such as lipopolysaccharide (LPS). Significantly, moderate doses of inflammogens produce new pathology in the brain and exacerbate or accelerate features of disease when superimposed upon existing pathology or in the context of genetic predisposition. It is now apparent in multiple chronic disease states, and in ageing, that microglia are primed by prior pathology, or by genetic predisposition, to respond more vigorously to subsequent inflammatory stimulation, thus transforming an adaptive CNS inflammatory response to systemic inflammation, into one that has deleterious consequences for the individual. In this review, the preclinical and clinical evidence supporting a significant role for systemic inflammation in chronic neurodegenerative diseases will be discussed. Mechanisms by which microglia might effect neuronal damage and dysfunction, as a consequence of systemic stimulation, will be highlighted. Copyright © 2012 Wiley Periodicals, Inc.
                Bookmark

                Author and article information

                Contributors
                +49 (391) 6713329 , ildikodunay@gmail.com
                Journal
                Acta Neuropathol Commun
                Acta Neuropathol Commun
                Acta Neuropathologica Communications
                BioMed Central (London )
                2051-5960
                16 March 2016
                16 March 2016
                2016
                : 4
                : 25
                Affiliations
                [ ]Institute for Medical Microbiology and Hospital Hygiene, University of Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
                [ ]Institute for Molecular and Clinical Immunology, University of Magdeburg, Magdeburg, Germany
                [ ]Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
                [ ]Department of Translational Inflammation Research, Institute of Experimental Internal Medicine, University of Magdeburg, Magdeburg, Germany
                [ ]Helmholtz Centre for Infection Research, Braunschweig, Germany
                [ ]Genetic Engineering and Biotechnology News, New York, USA
                [ ]Center for Behavioral Brain Sciences (CBBS), University of Magdeburg, Magdeburg, Germany
                [ ]Department of Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany
                [ ]Department of Psychiatry and Psychotherapy, Campus Mitte, Charité Universitätsmedizin, Berlin, Germany
                [ ]Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany
                [ ]Medical Faculty, University of Magdeburg, Magdeburg, Germany
                [ ]German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
                [ ]Neurogenetics, Leibniz Institute for Neurobiology, Magdeburg, Germany
                [ ]University of Lübeck (UzL), LIED, Lübeck, Germany
                [ ]Leibniz Institute of Plant Biochemistry (IPB), Halle, Germany
                Author information
                http://orcid.org/0000-0002-9900-8605
                Article
                293
                10.1186/s40478-016-0293-8
                4793516
                26984535
                24acb81b-2728-443b-ac4e-e0284cabd35e
                © Möhle et al. 2016

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 19 February 2016
                : 19 February 2016
                Funding
                Funded by: sfb 854
                Categories
                Research
                Custom metadata
                © The Author(s) 2016

                alzheimer’s disease,toxoplasma gondii,chronic infection,ly6chi monocytes,aβ clearance

                Comments

                Comment on this article