16
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Group A Streptococcus M1T1 Intracellular Infection of Primary Tonsil Epithelial Cells Dampens Levels of Secreted IL-8 Through the Action of SpyCEP

      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

          Streptococcus pyogenes (Group A Streptococcus; GAS) commonly causes pharyngitis in children and adults, with severe invasive disease and immune sequelae being an infrequent consequence. The ability of GAS to invade the host and establish infection likely involves subversion of host immune defenses. However, the signaling pathways and innate immune responses of epithelial cells to GAS are not well-understood. In this study, we utilized RNAseq to characterize the inflammatory responses of primary human tonsil epithelial (TEpi) cells to infection with the laboratory-adapted M6 strain JRS4 and the M1T1 clinical isolate 5448. Both strains induced the expression of genes encoding a wide range of inflammatory mediators, including IL-8. Pathway analysis revealed differentially expressed genes between mock and JRS4- or 5448-infected TEpi cells were enriched in transcription factor networks that regulate IL-8 expression, such as AP-1, ATF-2, and NFAT. While JRS4 infection resulted in high levels of secreted IL-8, 5448 infection did not, suggesting that 5448 may post-transcriptionally dampen IL-8 production. Infection with 5448Δ cepA, an isogenic mutant lacking the IL-8 protease SpyCEP, resulted in IL-8 secretion levels comparable to JRS4 infection. Complementation of 5448Δ cepA and JRS4 with a plasmid encoding 5448-derived SpyCEP significantly reduced IL-8 secretion by TEpi cells. Our results suggest that intracellular infection with the pathogenic GAS M1T1 clone induces a strong pro-inflammatory response in primary tonsil epithelial cells, but modulates this host response by selectively degrading the neutrophil-recruiting chemokine IL-8 to benefit infection.

          Related collections

          Most cited references49

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

          Transcription factors of the NFAT family: regulation and function.

          As targets for the immunosuppressive drugs cyclosporin A and FK506, transcription factors of the NFAT (nuclear factor of activated T cells) family have been the focus of much attention. NFAT proteins, which are expressed in most immune-system cells, play a pivotal role in the transcription of cytokine genes and other genes critical for the immune response. The activity of NFAT proteins is tightly regulated by the calcium/calmodulin-dependent phosphatase calcineurin, a primary target for inhibition by cyclosporin A and FK506. Calcineurin controls the translocation of NFAT proteins from the cytoplasm to the nucleus of activated cells by interacting with an N-terminal regulatory domain conserved in the NFAT family. The DNA-binding domains of NFAT proteins resemble those of Rel-family proteins, and Rel and NFAT proteins show some overlap in their ability to bind to certain regulatory elements in cytokine genes. NFAT is also notable for its ability to bind cooperatively with transcription factors of the AP-1 (Fos/Jun) family to composite NFAT:AP-1 sites, found in the regulatory regions of many genes that are inducibly transcribed by immune-system cells. This review discusses recent data on the diversity of the NFAT family of transcription factors, the regulation of NFAT proteins within cells, and the cooperation of NFAT proteins with other transcription factors to regulate the expression of inducible genes.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Disease manifestations and pathogenic mechanisms of group a Streptococcus.

            Streptococcus pyogenes, also known as group A Streptococcus (GAS), causes mild human infections such as pharyngitis and impetigo and serious infections such as necrotizing fasciitis and streptococcal toxic shock syndrome. Furthermore, repeated GAS infections may trigger autoimmune diseases, including acute poststreptococcal glomerulonephritis, acute rheumatic fever, and rheumatic heart disease. Combined, these diseases account for over half a million deaths per year globally. Genomic and molecular analyses have now characterized a large number of GAS virulence determinants, many of which exhibit overlap and redundancy in the processes of adhesion and colonization, innate immune resistance, and the capacity to facilitate tissue barrier degradation and spread within the human host. This improved understanding of the contribution of individual virulence determinants to the disease process has led to the formulation of models of GAS disease progression, which may lead to better treatment and intervention strategies. While GAS remains sensitive to all penicillins and cephalosporins, rising resistance to other antibiotics used in disease treatment is an increasing worldwide concern. Several GAS vaccine formulations that elicit protective immunity in animal models have shown promise in nonhuman primate and early-stage human trials. The development of a safe and efficacious commercial human vaccine for the prophylaxis of GAS disease remains a high priority.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              From RNA-seq reads to differential expression results

              Many methods and tools are available for preprocessing high-throughput RNA sequencing data and detecting differential expression.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Cell Infect Microbiol
                Front Cell Infect Microbiol
                Front. Cell. Infect. Microbiol.
                Frontiers in Cellular and Infection Microbiology
                Frontiers Media S.A.
                2235-2988
                17 May 2018
                2018
                : 8
                : 160
                Affiliations
                [1] 1School of Chemistry and Molecular Biosciences, University of Queensland , Brisbane, QLD, Australia
                [2] 2Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia , Perth, WA, Australia
                [3] 3Australian Institute for Bioengineering and Nanotechnology, University of Queensland , Brisbane, QLD, Australia
                [4] 4Australian Infectious Diseases Research Centre, University of Queensland , Brisbane, QLD, Australia
                [5] 5Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego , La Jolla, CA, United States
                [6] 6Centre for Stem Cell Systems, University of Melbourne , Melbourne, VIC, Australia
                [7] 7Institute for Molecular Bioscience and IMB Centre for Inflammation and Disease Research, University of Queensland , Brisbane, QLD, Australia
                Author notes

                Edited by: Kimberly Kline, Nanyang Technological University, Singapore

                Reviewed by: Jason W. Rosch, St. Jude Children's Research Hospital, United States; Nayeli Alva-Murillo, Universidad de Guanajuato, Mexico

                *Correspondence: Mark J. Walker mark.walker@ 123456uq.edu.au
                Article
                10.3389/fcimb.2018.00160
                5966554
                29868516
                41b06687-7bf7-4617-8b74-82644f791db1
                Copyright © 2018 Soderholm, Barnett, Korn, Rivera-Hernandez, Seymour, Schulz, Nizet, Wells, Sweet and Walker.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 19 February 2018
                : 26 April 2018
                Page count
                Figures: 4, Tables: 0, Equations: 0, References: 67, Pages: 13, Words: 10691
                Funding
                Funded by: National Health and Medical Research Council 10.13039/501100000925
                Award ID: APP1131932
                Award ID: APP1107914
                Award ID: APP1102621
                Categories
                Microbiology
                Original Research

                Infectious disease & Microbiology
                scpc,prts,il-8 protease,intracellular infection,streptococcus pyogenes

                Comments

                Comment on this article