15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Virus-induced inflammasome activation is suppressed by prostaglandin D 2/DP1 signaling

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      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.

          Significance

          Inflammatory responses to viral infections must be optimized to clear the pathogen without tissue damage. Inflammasomes comprise an important component of the innate immune response. Inflammasome activity must be carefully controlled to prevent a hyperinflammatory response, especially in brain infections. Here we identify a host factor, PYDC3, that is dependent upon prostaglandin D2 (PGD 2) and IFN-I signaling and is required to modulate inflammasome activation. After infection, inflammasome activation and expression of a downstream proinflammatory cytokine, IL-1β, were increased in mice deficient in PGD 2 signaling, decreasing survival. Excess mortality was reversed by IL-1β receptor blockade. These results define a consequence of prostaglandin signaling and shed light on prostaglandin–inflammasome interactions, which modulate excessive inflammation and tissue damage in the virus-infected brain.

          Abstract

          Prostaglandin D2 (PGD 2), an eicosanoid with both pro- and anti-inflammatory properties, is the most abundantly expressed prostaglandin in the brain. Here we show that PGD 2 signaling through the D-prostanoid receptor 1 (DP1) receptor is necessary for optimal microglia/macrophage activation and IFN expression after infection with a neurotropic coronavirus. Genome-wide expression analyses indicated that PGD 2/DP1 signaling is required for up-regulation of a putative inflammasome inhibitor, PYDC3, in CD11b + cells in the CNS of infected mice. Our results also demonstrated that, in addition to PGD 2/DP1 signaling, type 1 IFN (IFN-I) signaling is required for PYDC3 expression. In the absence of Pydc3 up-regulation, IL-1β expression and, subsequently, mortality were increased in infected DP1 −/− mice. Notably, survival was enhanced by IL1 receptor blockade, indicating that the effects of the absence of DP1 signaling on clinical outcomes were mediated, at least in part, by inflammasomes. Using bone marrow-derived macrophages in vitro, we confirmed that PYDC3 expression is dependent upon DP1 signaling and that IFN priming is critical for PYDC3 up-regulation. In addition, Pydc3 silencing or overexpression augmented or diminished IL-1β secretion, respectively. Furthermore, DP1 signaling in human macrophages also resulted in the up-regulation of a putative functional analog, POP3, suggesting that PGD 2 similarly modulates inflammasomes in human cells. These findings demonstrate a previously undescribed role for prostaglandin signaling in preventing excessive inflammasome activation and, together with previously published results, suggest that eicosanoids and inflammasomes are reciprocally regulated.

          Related collections

          Most cited references38

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

          Interleukin-1 and neuronal injury.

          Interleukin-1 is a pro-inflammatory cytokine that has numerous biological effects, including activation of many inflammatory processes (through activation of T cells, for example), induction of expression of acute-phase proteins, an important function in neuroimmune responses and direct effects on the brain itself. There is now extensive evidence to support the direct involvement of interleukin-1 in the neuronal injury that occurs in both acute and chronic neurodegenerative disorders. This article discusses the key evidence of a role for interleukin-1 in acute neurodegeneration - for example, stroke and brain trauma - and provides a rationale for targeting the interleukin-1 system as a therapeutic strategy.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Programmed necrosis in inflammation: Toward identification of the effector molecules.

            Until recently, programmed cell death was conceived of as a single set of molecular pathways. We now know of several distinct sets of death-inducing mechanisms that lead to differing cell-death processes. In one of them--apoptosis--the dying cell affects others minimally. In contrast, programmed necrotic cell death causes release of immunostimulatory intracellular components after cell-membrane rupture. Defining the in vivo relevance of necrotic death is hampered because the molecules initiating it [such as receptor-interacting protein kinase-1 (RIPK1), RIPK3, or caspase-1] also serve other functions. Proteins that participate in late events in two forms of programmed necrosis [mixed lineage kinase domain-like protein (MLKL) in necroptosis and gasdermin-D in pyroptosis] were recently discovered, bringing us closer to identifying molecules that strictly serve in death mediation, thereby providing probes for better assessing its role in inflammation.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Prostaglandin D2 Selectively Induces Chemotaxis in T Helper Type 2 Cells, Eosinophils, and Basophils via Seven-Transmembrane Receptor Crth2

              Prostaglandin (PG)D2, which has long been implicated in allergic diseases, is currently considered to elicit its biological actions through the DP receptor (DP). Involvement of DP in the formation of allergic asthma was recently demonstrated with DP-deficient mice. However, proinflammatory functions of PGD2 cannot be explained by DP alone. We show here that a seven-transmembrane receptor, CRTH2, which is preferentially expressed in T helper type 2 (Th2) cells, eosinophils, and basophils in humans, serves as the novel receptor for PGD2. In response to PGD2, CRTH2 induces intracellular Ca2+ mobilization and chemotaxis in Th2 cells in a Gαi-dependent manner. In addition, CRTH2, but not DP, mediates PGD2-dependent cell migration of blood eosinophils and basophils. Thus, PGD2 is likely involved in multiple aspects of allergic inflammation through its dual receptor systems, DP and CRTH2.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                3 July 2017
                19 June 2017
                : 114
                : 27
                : E5444-E5453
                Affiliations
                [1] aInterdisciplinary Program in Immunology, University of Iowa , Iowa City, IA 52242;
                [2] bDepartment of Microbiology, University of Iowa , Iowa City, IA 52242;
                [3] cIowa Institute of Human Genetics, University of Iowa , Iowa City, IA 52242;
                [4] dDepartment of Pathology, University of Iowa , Iowa City, IA 52242;
                [5] eDepartment of Internal Medicine, University of Iowa , Iowa City, IA 52242;
                [6] fDepartment of Pharmacology, Kyoto University Faculty of Medicine , Tokyo, Japan 606-8501
                Author notes
                2To whom correspondence should be addressed. Email: stanley-perlman@ 123456uiowa.edu .

                Edited by Diane E. Griffin, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, and approved May 31, 2017 (received for review March 10, 2017)

                Author contributions: R.V., A.R.F., S.P.K., F.S.S., and S.P. designed research; R.V., A.R.F., A.M.J., J.A., D.L.W., and M.G. performed research; S.N. contributed new reagents/analytic tools; R.V., R.S., D.K.M., and S.P. analyzed data; and R.V. and S.P. wrote the paper.

                1Present address: Department of Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048.

                Author information
                http://orcid.org/0000-0003-4213-2354
                Article
                PMC5502630 PMC5502630 5502630 201704099
                10.1073/pnas.1704099114
                5502630
                28630327
                f7ea579e-5e16-4ce1-853f-5832a5b01aff
                History
                Page count
                Pages: 10
                Funding
                Funded by: HHS | National Institutes of Health (NIH) 100000002
                Award ID: NS36592
                Funded by: HHS | National Institutes of Health (NIH) 100000002
                Award ID: AI118719
                Funded by: HHS | National Institutes of Health (NIH) 100000002
                Award ID: AI007485
                Funded by: HHS | National Institutes of Health (NIH) 100000002
                Award ID: P30CA086862
                Funded by: National Multiple Sclerosis Society 100000890
                Award ID: RG5340-A-7
                Categories
                PNAS Plus
                Biological Sciences
                Microbiology
                PNAS Plus

                coronavirus,inflammasomes,pyrin domain-only protein,encephalitis,prostaglandin D2

                Comments

                Comment on this article