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

      Identification of genes required for Mycobacterium abscessus growth in vivo with a prominent role of the ESX-4 locus

      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

          The coevolution of mycobacteria and amoebae seems to have contributed to shaping the virulence of nontuberculous mycobacteria in macrophages. We identified a pool of genes essential for the intracellular survival of Mycobacterium abscessus inside amoebae and macrophages and discovered a hot spot of transposon insertions within the orthologous ESX-4 T7SS locus. We generated a mutant with the deletion of a structural key ESX component, EccB 4. We demonstrate rupture of the phagosomal membrane only in the presence of an intact eccB 4 gene. These results suggest an unanticipated role of ESX-4 T7SS in governing the intracellular behavior of a mycobacterium. Because M. abscessus lacks ESX-1, it is tempting to speculate that ESX-4 operates as a surrogate for ESX-1 in M. tuberculosis.

          Abstract

          Mycobacterium abscessus, a rapidly growing mycobacterium (RGM) and an opportunistic human pathogen, is responsible for a wide spectrum of clinical manifestations ranging from pulmonary to skin and soft tissue infections. This intracellular organism can resist the bactericidal defense mechanisms of amoebae and macrophages, an ability that has not been observed in other RGM. M. abscessus can up-regulate several virulence factors during transient infection of amoebae, thereby becoming more virulent in subsequent respiratory infections in mice. Here, we sought to identify the M. abscessus genes required for replication within amoebae. To this end, we constructed and screened a transposon ( Tn) insertion library of an M. abscessus subsp ecies massiliense clinical isolate for attenuated clones. This approach identified five genes within the ESX-4 locus, which in M. abscessus encodes an ESX-4 type VII secretion system that exceptionally also includes the ESX conserved EccE component. To confirm the screening results and to get further insight into the contribution of ESX-4 to M. abscessus growth and survival in amoebae and macrophages, we generated a deletion mutant of eccB 4 that encodes a core structural element of ESX-4. This mutant was less efficient at blocking phagosomal acidification than its parental strain. Importantly, and in contrast to the wild-type strain, it also failed to damage phagosomes and showed reduced signs of phagosome-to-cytosol contact, as demonstrated by a combination of cellular and immunological assays. This study attributes an unexpected and genuine biological role to the underexplored mycobacterial ESX-4 system and its substrates.

          Related collections

          Most cited references44

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

          Microorganisms resistant to free-living amoebae.

          Free-living amoebae feed on bacteria, fungi, and algae. However, some microorganisms have evolved to become resistant to these protists. These amoeba-resistant microorganisms include established pathogens, such as Cryptococcus neoformans, Legionella spp., Chlamydophila pneumoniae, Mycobacterium avium, Listeria monocytogenes, Pseudomonas aeruginosa, and Francisella tularensis, and emerging pathogens, such as Bosea spp., Simkania negevensis, Parachlamydia acanthamoebae, and Legionella-like amoebal pathogens. Some of these amoeba-resistant bacteria (ARB) are lytic for their amoebal host, while others are considered endosymbionts, since a stable host-parasite ratio is maintained. Free-living amoebae represent an important reservoir of ARB and may, while encysted, protect the internalized bacteria from chlorine and other biocides. Free-living amoebae may act as a Trojan horse, bringing hidden ARB within the human "Troy," and may produce vesicles filled with ARB, increasing their transmission potential. Free-living amoebae may also play a role in the selection of virulence traits and in adaptation to survival in macrophages. Thus, intra-amoebal growth was found to enhance virulence, and similar mechanisms seem to be implicated in the survival of ARB in response to both amoebae and macrophages. Moreover, free-living amoebae represent a useful tool for the culture of some intracellular bacteria and new bacterial species that might be potential emerging pathogens.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Type VII secretion--mycobacteria show the way.

            Recent evidence shows that mycobacteria have developed novel and specialized secretion systems for the transport of extracellular proteins across their hydrophobic, and highly impermeable, cell wall. Strikingly, mycobacterial genomes encode up to five of these transport systems. Two of these systems, ESX-1 and ESX-5, are involved in virulence - they both affect the cell-to-cell migration of pathogenic mycobacteria. Here, we discuss this novel secretion pathway and consider variants that are present in various Gram-positive bacteria. Given the unique composition of this secretion system, and its general importance, we propose that, in line with the accepted nomenclature, it should be called type VII secretion.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mycobacterium tuberculosis Differentially Activates cGAS- and Inflammasome-Dependent Intracellular Immune Responses through ESX-1.

              Cytosolic detection of microbial products is essential for the initiation of an innate immune response against intracellular pathogens such as Mycobacterium tuberculosis (Mtb). During Mtb infection of macrophages, activation of cytosolic surveillance pathways is dependent on the mycobacterial ESX-1 secretion system and leads to type I interferon (IFN) and interleukin-1β (IL-1β) production. Whereas the inflammasome regulates IL-1β secretion, the receptor(s) responsible for the activation of type I IFNs has remained elusive. We demonstrate that the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) is essential for initiating an IFN response to Mtb infection. cGAS associates with Mtb DNA in the cytosol to stimulate cyclic GAMP (cGAMP) synthesis. Notably, activation of cGAS-dependent cytosolic host responses can be uncoupled from inflammasome activation by modulating the secretion of ESX-1 substrates. Our findings identify cGAS as an innate sensor of Mtb and provide insight into how ESX-1 controls the activation of specific intracellular recognition pathways.
                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
                30 January 2018
                17 January 2018
                : 115
                : 5
                : E1002-E1011
                Affiliations
                [1] a Université de Versailles Saint Quentin en Yvelines , INSERM UMR1173, 78000 Versailles, France;
                [2] bInstitut de Recherche en Infectiologie de Montpellier, Université de Montpellier , CNRS UMR 9004, 34293 Montpellier, France;
                [3] cUnité de Pathogénomique Mycobactérienne, Institut Pasteur , 75015 Paris, France;
                [4] dDepartment of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health , Boston, MA 02115;
                [5] eAssistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Ile de France Ouest , Ambroise Paré, Boulogne and Raymond Poincaré, 92380 Garches, France;
                [6] fLaboratoire de spectrométrie de masse protéomique, Institut Curie, Paris Science and Letters Research University , 75248 Paris, France;
                [7] gINSERM, Institut de Recherche en Infectiologie de Montpellier , 34293 Montpellier, France
                Author notes
                3To whom correspondence may be addressed. Email: jean-louis.herrmann@ 123456aphp.fr or fabienne.misguich@ 123456uvsq.fr .

                Edited by Marcel A. Behr, McGill International TB Centre, Montreal, Quebec, Canada, and accepted by Editorial Board Member Carl F. Nathan December 18, 2017 (received for review July 25, 2017)

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

                1Present address: Department of Biomedical Engineering and the Huck Institutes of the Life Sciences, Pennsylvania State University, State College, PA 16801.

                2Present address: Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, United Kingdom.

                Author information
                http://orcid.org/0000-0002-9111-8842
                http://orcid.org/0000-0003-2587-3863
                http://orcid.org/0000-0002-7571-6519
                Article
                PMC5798338 PMC5798338 5798338 201713195
                10.1073/pnas.1713195115
                5798338
                29343644
                c6e6ef13-b58b-474e-b464-5aac7cbdd6ec
                Copyright @ 2018

                Published under the PNAS license.

                History
                Page count
                Pages: 10
                Categories
                PNAS Plus
                Biological Sciences
                Microbiology
                PNAS Plus

                survival,TVIISS-ESX4, M. abscessus
                survival, TVIISS-ESX4, M. abscessus

                Comments

                Comment on this article