14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Group A Streptococcus Induces LAPosomes via SLO/β1 Integrin/NOX2/ROS Pathway in Endothelial Cells That Are Ineffective in Bacterial Killing and Suppress Xenophagy

      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

          Our previous reports showed that the LC3-associated GAS-containing single membrane vacuoles are inefficient for bacterial clearance in endothelial cells, which may result in bacteremia. However, the characteristics and the induction mechanisms of these LC3-positive vacuoles are still largely unknown. Here we provide the first evidence that these LC3-positive GAS-containing single membrane compartments appear to be LAPosomes, which are induced by NOX2 and ROS. Through NOX2- and ROS-mediated signaling, GAS preferentially induces LAP and inhibits bacteriostatic xenophagy in endothelial cells. We also provide the first demonstration that β1 integrin acts as the receptor for LAP induction through GAS-produced SLO stimulation in endothelial cells. Our findings reveal the underlying mechanisms of LAP induction and autophagy evasion for GAS multiplication in endothelial cells.

          ABSTRACT

          Group A streptococcus (GAS) is an important human pathogen which can cause fatal diseases after invasion into the bloodstream. Although antibiotics and immune surveillance are the main defenses against GAS infection, GAS utilizes internalization into cells as a major immune evasion strategy. Our previous findings revealed that light chain 3 (LC3)-associated single membrane GAS-containing vacuoles in endothelial cells are compromised for bacterial clearance due to insufficient acidification after fusion with lysosomes. However, the characteristics and the activation mechanisms of these LC3-positive compartments are still largely unknown. In the present study, we demonstrated that the LC3-positive GAS is surrounded by single membrane and colocalizes with NADPH oxidase 2 (NOX2) complex but without ULK1, which are characteristics of LC3-associated phagocytosis (LAP). Inhibition of NOX2 or reactive oxygen species (ROS) significantly reduces GAS multiplication and enhances autolysosome acidification in endothelial cells through converting LAP to conventional xenophagy, which is revealed by enhancement of ULK1 recruitment, attenuation of p70s6k phosphorylation, and formation of the isolation membrane. We also clarify that the inactivation of mTORC1, which is the initiation signal of autophagy, is inhibited by NOX2- and ROS-activated phosphatidylinositol 3-kinase (PI3K)/AKT and MEK/extracellular signal-regulated kinase (ERK) pathways. In addition, streptolysin O (SLO) of GAS is identified as a crucial inducer of ROS for β1 integrin-mediated LAP induction. After downregulation of β1 integrin, GAS multiplication is reduced, accompanied with LAP inhibition and xenophagy induction. These results demonstrate that GAS infection preferentially induces ineffective LAP to evade xenophagic killing in endothelial cells through the SLO/β1 integrin/NOX2/ROS pathway.

          Related collections

          Most cited references46

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

          The global burden of group A streptococcal diseases.

          The global burden of disease caused by group A streptococcus (GAS) is not known. We review recent population-based data to estimate the burden of GAS diseases and highlight deficiencies in the available data. We estimate that there are at least 517,000 deaths each year due to severe GAS diseases (eg, acute rheumatic fever, rheumatic heart disease, post-streptococcal glomerulonephritis, and invasive infections). The prevalence of severe GAS disease is at least 18.1 million cases, with 1.78 million new cases each year. The greatest burden is due to rheumatic heart disease, with a prevalence of at least 15.6 million cases, with 282,000 new cases and 233,000 deaths each year. The burden of invasive GAS diseases is unexpectedly high, with at least 663,000 new cases and 163,000 deaths each year. In addition, there are more than 111 million prevalent cases of GAS pyoderma, and over 616 million incident cases per year of GAS pharyngitis. Epidemiological data from developing countries for most diseases is poor. On a global scale, GAS is an important cause of morbidity and mortality. These data emphasise the need to reinforce current control strategies, develop new primary prevention strategies, and collect better data from developing countries.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The TBK1 adaptor and autophagy receptor NDP52 restricts the proliferation of ubiquitin-coated bacteria.

            Cell-autonomous innate immune responses against bacteria attempting to colonize the cytosol of mammalian cells are incompletely understood. Polyubiquitylated proteins can accumulate on the surface of such bacteria, and bacterial growth is restricted by Tank-binding kinase (TBK1). Here we show that NDP52, not previously known to contribute to innate immunity, recognizes ubiquitin-coated Salmonella enterica in human cells and, by binding the adaptor proteins Nap1 and Sintbad, recruits TBK1. Knockdown of NDP52 and TBK1 facilitated bacterial proliferation and increased the number of cells containing ubiquitin-coated salmonella. NDP52 also recruited LC3, an autophagosomal marker, and knockdown of NDP52 impaired autophagy of salmonella. We conclude that human cells utilize the ubiquitin system and NDP52 to activate autophagy against bacteria attempting to colonize their cytosol.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Autophagy defends cells against invading group A Streptococcus.

              We found that the autophagic machinery could effectively eliminate pathogenic group A Streptococcus (GAS) within nonphagocytic cells. After escaping from endosomes into the cytoplasm, GAS became enveloped by autophagosome-like compartments and were killed upon fusion of these compartments with lysosomes. In autophagy-deficient Atg5-/- cells, GAS survived, multiplied, and were released from the cells. Thus, the autophagic machinery can act as an innate defense system against invading pathogens.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Role: Solicited external reviewer
                Role: Solicited external reviewer
                Journal
                mBio
                MBio
                mbio
                mbio
                mBio
                mBio
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                2150-7511
                1 October 2019
                Sep-Oct 2019
                : 10
                : 5
                : e02148-19
                Affiliations
                [a ]Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang Ming University, Taipei, Taiwan
                [b ]Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
                [c ]Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
                [d ]School of Medicine, I-Shou University, Kaohsiung, Taiwan
                [e ]Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka, Japan
                [f ]Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
                [g ]Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
                [h ]Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
                [i ]Department of Microbiology & Immunology, Dalhousie University, Halifax, Canada
                [j ]Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei, Taiwan
                [k ]Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
                [l ]School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
                [m ]Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
                College of Veterinary Medicine, Cornell University
                Montana State University-Bozeman
                Chang Gung University
                Author notes
                Address correspondence to Jiunn-Jong Wu, jjwu1019@ 123456ym.edu.tw , or Yee-Shin Lin, yslin1@ 123456mail.ncku.edu.tw .
                Author information
                https://orcid.org/0000-0003-3581-7961
                Article
                mBio02148-19
                10.1128/mBio.02148-19
                6775456
                31575768
                ba4a155b-1022-45f7-92fe-96dbaab10b8f
                Copyright © 2019 Cheng et al.

                This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

                History
                : 15 August 2019
                : 29 August 2019
                Page count
                Figures: 6, Tables: 0, Equations: 0, References: 58, Pages: 15, Words: 8916
                Funding
                Funded by: Ministry of Science and Technology, Taiwan (MOST), https://doi.org/10.13039/501100004663;
                Award ID: 107-2320-B-006-050
                Award ID: 108-2320-B-006-011
                Award Recipient :
                Funded by: Ministry of Science and Technology, Taiwan (MOST), https://doi.org/10.13039/501100004663;
                Award ID: 107-2320-B-010-021
                Award ID: 108-2320-B-010-004
                Award Recipient :
                Categories
                Research Article
                Host-Microbe Biology
                Custom metadata
                September/October 2019

                Life sciences
                group a streptococcus,lc3-associated phagocytosis (lap),xenophagy,reactive oxygen species (ros),endothelial cells

                Comments

                Comment on this article