29
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      DNA-induced 2′3′-cGAMP enhances haplotype-specific human STING cleavage by dengue protease

      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.

          Significance

          Dengue virus (DENV) antagonizes the DNA sensing cGAS-STING pathway to subvert innate immunity, but how DENV protease-mediated human STING cleavage contributes to DENV pathogenesis remains obscure. Here, we found that STING haplotype frequency varies among different subhuman populations, and different haplotypes respond differently to DENV protease. The cleavage of a DENV protease-sensitive STING can be further enhanced by coculture with neighboring cells producing 2′3′-cGAMP, either by DNA transfection of cGAS or by reactivating Epstein–Barr virus from latent infection. Thus, DENV infection trims down human STING-mediated innate immunity in a haplotype-specific manner. The genetic background of host STING and bystander coinfection of pathogens triggering 2′3′-cGAMP production may be the missing link between STING cleavage and DENV pathogenesis.

          Abstract

          The cytosolic DNA sensor cGMP-AMP synthase (cGAS) synthesizes the noncanonical cyclic dinucleotide 2′3′-cGAMP to activate the adaptor protein stimulator of IFN genes (STING), thus awakening host immunity in response to DNA pathogen infection. However, dengue virus (DENV), an RNA virus without a DNA stage in its life cycle, also manipulates cGAS-STING–mediated innate immunity by proteolytic degradation of STING. Here, we found that the sensitivity of STING to DENV protease varied with different human STING haplotypes. Exogenous DNA further enhanced DENV protease’s ability to interact and cleave protease-sensitive STING. DNA-enhanced STING cleavage was reduced in cGAS-knockdown cells and triggered by the cGAS product 2′3′-cGAMP. The source of DNA may not be endogenous mitochondrial DNA but rather exogenous reactivated viral DNA. Cells producing 2′3′-cGAMP by overexpressing cGAS or with DNA virus reactivation enhanced STING cleavage in neighboring cells harboring DENV protease. DENV infection reduced host innate immunity in cells with the protease-sensitive STING haplotype, whose homozygote genotype frequency was found significantly reduced in Taiwanese people with dengue fever. Therefore, the human STING genetic background and DNA pathogen coinfection may be the missing links contributing to DENV pathogenesis.

          Related collections

          Most cited references23

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

          Structural basis of STING binding with and phosphorylation by TBK1

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

            Atg9a controls dsDNA-driven dynamic translocation of STING and the innate immune response.

            Microbial nucleic acids are critical for the induction of innate immune responses, a host defense mechanism against infection by microbes. Recent studies have indicated that double-stranded DNA (dsDNA) induces potent innate immune responses via the induction of type I IFN (IFN) and IFN-inducible genes. However, the regulatory mechanisms underlying dsDNA-triggered signaling are not fully understood. Here we show that the translocation and assembly of the essential signal transducers, stimulator of IFN genes (STING) and TANK-binding kinase 1 (TBK1), are required for dsDNA-triggered innate immune responses. After sensing dsDNA, STING moves from the endoplasmic reticulum (ER) to the Golgi apparatus and finally reaches the cytoplasmic punctate structures to assemble with TBK1. The addition of an ER-retention signal to the C terminus of STING dampens its ability to induce antiviral responses. We also show that STING co-localizes with the autophagy proteins, microtubule-associated protein 1 light chain 3 (LC3) and autophagy-related gene 9a (Atg9a), after dsDNA stimulation. The loss of Atg9a, but not that of another autophagy-related gene (Atg7), greatly enhances the assembly of STING and TBK1 by dsDNA, leading to aberrant activation of the innate immune response. Hence Atg9a functions as a regulator of innate immunity following dsDNA stimulation as well as an essential autophagy protein. These results demonstrate that dynamic membrane traffic mediates the sequential translocation and assembly of STING, both of which are essential processes required for maximal activation of the innate immune response triggered by dsDNA.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found

              Global Spread and Persistence of Dengue

              Dengue is a spectrum of disease caused by four serotypes of the most prevalent arthropod-borne virus affecting humans today, and its incidence has increased dramatically in the past 50 years. Due in part to population growth and uncontrolled urbanization in tropical and subtropical countries, breeding sites for the mosquitoes that transmit dengue virus have proliferated, and successful vector control has proven problematic. Dengue viruses have evolved rapidly as they have spread worldwide, and genotypes associated with increased virulence have expanded from South and Southeast Asia into the Pacific and the Americas. This review explores the human, mosquito, and viral factors that contribute to the global spread and persistence of dengue, as well as the interaction between the three spheres, in the context of ecological and climate changes. What is known, as well as gaps in knowledge, is emphasized in light of future prospects for control and prevention of this pandemic disease.
                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
                7 July 2020
                23 June 2020
                23 June 2020
                : 117
                : 27
                : 15947-15954
                Affiliations
                [1] aNational Institute of Infectious Diseases and Vaccinology, National Health Research Institutes , Miaoli 350, Taiwan;
                [2] bDepartment of Microbiology and Immunology, National Cheng Kung University , 701 Tainan, Taiwan;
                [3] cDepartment of Pediatrics, National Cheng Kung University , 701 Tainan, Taiwan;
                [4] dInstitute of Molecular Medicine, National Cheng Kung University , 701 Tainan, Taiwan;
                [5] eInstitute of Biomedical Sciences , Academia Sinica, 115 Taipei, Taiwan;
                [6] fResearch Center for Emerging Viruses, China Medical University Hospital , 404 Taichung, Taiwan;
                [7] gInstitute of Molecular Biology , Academia Sinica, 115 Taipei, Taiwan;
                [8] hGraduate Institute of Integrated Medicine, China Medical University , 404 Taichung, Taiwan
                Author notes
                2To whom correspondence may be addressed. Email: epitope@ 123456nhri.edu.tw .

                Edited by Adolfo Garcia-Sastre, Icahn School of Medicine at Mount Sinai, New York, NY, and approved May 27, 2020 (received for review December 18, 2019)

                Author contributions: Y.C. and C.-Y.Y. designed research; C.-I.S., Y.-T.K., C.-C.C., and C.-Y.Y. performed research; T.-S.H., H.S.S., and Y.-H.L. contributed new reagents/analytic tools; Y.-L.L., M.M.C.L., and C.-Y.Y. analyzed data; and Y.-L.L., M.M.C.L., and C.-Y.Y. wrote the paper.

                1C.-I.S. and Y.-T.K. contributed equally to this work.

                Author information
                https://orcid.org/0000-0003-2951-6679
                https://orcid.org/0000-0002-3661-4448
                https://orcid.org/0000-0002-3603-868X
                Article
                201922243
                10.1073/pnas.1922243117
                7354927
                32576686
                7437ae03-270a-4fb9-b3c3-f158323c4234
                Copyright © 2020 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution License 4.0 (CC BY).

                History
                Page count
                Pages: 8
                Funding
                Funded by: Ministry of Science and Technology, Taiwan (MOST) 501100004663
                Award ID: MOST106-2320-B-400-031-MY3
                Award Recipient : Yi-Ling Lin Award Recipient : Chia-Yi Yu
                Funded by: National Health Research Institutes (NHRI) 501100004737
                Award ID: IV-108-PP-27 and MR-108-GP-06
                Award Recipient : Chia-Yi Yu
                Funded by: Ministry of Science and Technology, Taiwan (MOST) 501100004663
                Award ID: 108-2321-B-001-011
                Award Recipient : Yi-Ling Lin Award Recipient : Chia-Yi Yu
                Categories
                Biological Sciences
                Microbiology

                sting,denv protease,snp,2′3′-cgamp,cgas
                sting, denv protease, snp, 2′3′-cgamp, cgas

                Comments

                Comment on this article